WO2008009132A1 - Tissue rejection - Google Patents

Tissue rejection Download PDF

Info

Publication number
WO2008009132A1
WO2008009132A1 PCT/CA2007/001295 CA2007001295W WO2008009132A1 WO 2008009132 A1 WO2008009132 A1 WO 2008009132A1 CA 2007001295 W CA2007001295 W CA 2007001295W WO 2008009132 A1 WO2008009132 A1 WO 2008009132A1
Authority
WO
WIPO (PCT)
Prior art keywords
tissue
nucleic acid
profile
transplanted
injury
Prior art date
Application number
PCT/CA2007/001295
Other languages
French (fr)
Inventor
Philip F. Halloran
Original Assignee
The Governors Of The University Of Alberta
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Governors Of The University Of Alberta filed Critical The Governors Of The University Of Alberta
Priority to US12/374,639 priority Critical patent/US20090176656A1/en
Priority to EP07784962A priority patent/EP2049713A4/en
Publication of WO2008009132A1 publication Critical patent/WO2008009132A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/16Primer sets for multiplex assays
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/34Genitourinary disorders
    • G01N2800/347Renal failures; Glomerular diseases; Tubulointerstitial diseases, e.g. nephritic syndrome, glomerulonephritis; Renovascular diseases, e.g. renal artery occlusion, nephropathy

Definitions

  • tissue injury such as tissue injury that may occur with organ transplant rejection (alloimmune injury) or non-alloimmune injury.
  • tissue rejection relates to methods and materials involved in detecting tissue rejection.
  • tissue rejection and tissue injury that may be due to alloimmune or non-alloimmune events is a concern for any recipient of transplanted tissue. If a clinician is able to recognize early signs of tissue rejection, anti-rejection drugs and other medication often can be used to reverse tissue rejection and manage injury. Further, understanding molecular mechanisms of injury and rejection will lead to development of improved diagnostics and therapeutics.
  • kidney transplantation the renal tubular epithelium is a key target of rejection. Changes in the epithelium have diagnostic significance in T cell mediated renal allograft rejection (TCMR). Entry of mononuclear inflammatory cells into the renal tubular epithelium during TCMR (Racusen et al. (1999) Kidney Int. 55:713-723) is associated with deterioration of renal function (Solez et al. (1993) Kidney Int. 43:1058-1067; and Solez et al.
  • TCMR T cell mediated renal allograft rejection
  • Kidney Int. 44:411-422 Tubulitis, associated with interstitial infiltration by mononuclear cells, is the principal lesion used to diagnose TCMR using the Banff schema (a pathology diagnostic system; Racusen et al. ⁇ supra). Kidneys also can be injured by antibody-mediated rejection
  • tissue injury such as injury inherent in an organ that is transplanted or is to be transplanted, or injury that occurs with organ transplantation (e.g., alloimmune injury associated with rejection, or non-alloimune injury that can occur, for example, during surgery).
  • tissue injury e.g., tissue injury due to kidney rejection
  • assessment of a mammal's probability of rejecting tissue such as a transplanted organ.
  • This document also relates to methods and materials involved in assessment of tissue quality and performance (e.g., assessment of donor organs for transplantation, prediction of whether an organ is at increased risk for developing delayed graft function (DGF) following transplantation, and assessment of transplanted organs and their potential to recover from alloimmune or non-alloimmune injury).
  • assessment of tissue quality and performance e.g., assessment of donor organs for transplantation, prediction of whether an organ is at increased risk for developing delayed graft function (DGF) following transplantation, and assessment of transplanted organs and their potential to recover from alloimmune or non-alloimmune injury.
  • DGF delayed graft function
  • tissue injury can be detected at a time point prior to the emergence of any visually-observable, histological sign of injury (e.g., in kidney tissue, tubulitis, loss of epithelial mass, marked reduction of E-cadherin and Ksp-cadherin, and redistribution to the apical membrane).
  • histological sign of injury e.g., in kidney tissue, tubulitis, loss of epithelial mass, marked reduction of E-cadherin and Ksp-cadherin, and redistribution to the apical membrane.
  • expression levels of "injury-and-repair induced transcripts” IRITs
  • “not in isografts injury-and-repair induced transcripts” NIRITs
  • GSTs gamma-interferon suppressed transcripts
  • CISTs class I suppressed transcripts
  • Solute carriers (Slcs) and renal transcripts (RTs) listed in Tables 1-4 can be assessed to determine whether or not tissue is injured, or to distinguish transplanted tissue that is injured from transplanted tissue that is not injured.
  • the expression level of gene profiles that significantly correlate with the sets referred to in Tables 1-14 can be assessed to determine whether or not tissue is injured, or to distinguish transplanted tissue that is injured from transplanted tissue that is not injured.
  • nucleic acid arrays that can be used to diagnose tissue injury in a mammal.
  • Such arrays can, for example, allow clinicians to diagnose injury in a donor biopsy, diagnose tissue injury in a transplanted organ, or determine the potential for recovery of organ function in a transplanted organ, based on determination of the expression levels of nucleic acids that are differentially expressed in injured and/or rejected tissue as compared to control tissue that is not injured or rejected.
  • the differential expression of such nucleic acids can be detected in injured tissue prior to the emergence of visually-observable, histological signs of tissue injury or rejection, allowing for early diagnosis of patients having injured transplanted tissue.
  • Such diagnosis can help clinicians determine appropriate treatments for those patients. For example, a clinician who diagnoses a patient as having injured transplanted tissue can treat that patient with medication that suppresses tissue rejection and thus injury (e.g., immunosuppressants). In addition, better therapeutics can be developed that will treat or manage injury events.
  • this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having an injury and repair profile, wherein the presence of the cells indicates that the tissue is injured.
  • the mammal can be a human.
  • the tissue can be from a biopsy.
  • the tissue can be kidney tissue.
  • the tissue can be tissue to be transplanted into a recipient.
  • the tissue can be tissue that has been transplanted into a recipient.
  • the determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
  • this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a not-in-isografts injury and repair profile, wherein the presence of the cells indicates that the tissue is injured.
  • the mammal can be a human.
  • the tissue can be from a biopsy.
  • the tissue can be kidney tissue.
  • the tissue can be tissue to be transplanted into a recipient.
  • the tissue can be tissue that has been transplanted into a recipient.
  • the determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
  • this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a gamma interferon (IFN-K) suppressed profile, wherein the presence of the cells indicates that the tissue is injured.
  • the mammal can be a human.
  • the tissue can be from a biopsy.
  • the tissue can be kidney tissue.
  • the tissue can be tissue to be transplanted into a recipient.
  • the tissue can be tissue that has been transplanted into a recipient.
  • the determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
  • this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a class I suppressed profile, wherein the presence of the cells indicates that the tissue is injured.
  • the mammal can be a human.
  • the tissue can be from a biopsy.
  • the tissue can be kidney tissue.
  • the tissue can be tissue to be transplanted into a recipient.
  • the tissue can be tissue that has been transplanted into a recipient.
  • the determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
  • this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a renal transcript (RT) profile, wherein the presence of the cells indicates that the tissue is injured.
  • the mammal can be a human.
  • the tissue can be from a biopsy.
  • the tissue can be kidney tissue.
  • the tissue can be tissue to be transplanted into a recipient.
  • the tissue can be tissue that has been transplanted into a recipient.
  • the determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
  • this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a solute carrier (SIc) profile, wherein the presence of the cells indicates that the tissue is injured.
  • the mammal can be a human.
  • the tissue can be from a biopsy.
  • the tissue can be kidney tissue.
  • the tissue can be tissue to be transplanted into a recipient.
  • the tissue can be tissue that has been transplanted into a recipient.
  • the determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
  • This document also features a method for assessing whether a tissue is at risk for delayed graft function (DGF), wherein the method comprises determining whether or not a tissue contains cells having an injury and repair profile, a non-in-isografts injury and repair profile, an IFN-K suppressed profile, a class I suppressed profile, a RT profile, or a SIc profile, wherein the presence of the cells indicates that the tissue is at risk for DGF.
  • the mammal can be a human.
  • the tissue can be from a biopsy.
  • the tissue can be kidney tissue.
  • the tissue can be tissue to be transplanted into a recipient.
  • the tissue can be tissue that has been transplanted into a recipient.
  • the determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
  • this document features a method for predicting whether a transplanted tissue will recover from injury, wherein the method comprises determining whether or not a tissue contains cells having an injury and repair profile, a non-in- isografts injury and repair profile, an IFN-K suppressed profile, a class I suppressed profile, a RT profile, or a SIc profile, wherein the presence of the cells indicates that the tissue is not likely to recover from injury.
  • the mammal can be a human.
  • the tissue can be from a biopsy.
  • the tissue can be kidney tissue.
  • the tissue can be tissue to be transplanted into a recipient.
  • the tissue can be tissue that has been transplanted into a recipient.
  • the determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
  • this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having an injury and repair correlated profile or an SIc correlated profile, wherein the presence of the cells indicates that the tissue is injured.
  • the mammal can be a human.
  • the tissue can be from a biopsy.
  • the tissue can be kidney tissue.
  • the tissue can be tissue to be transplanted into a recipient.
  • the tissue can be tissue that has been transplanted into a recipient.
  • the determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
  • This document also features a method for detecting tissue injury, comprising determining whether or not a tissue contains cells having increased activity of biochemical pathways that correlate with an injury and repair profile, with an SIc profile, with a non-in-isografts injury and repair profile, with a gamma interferon suppressed profile, with a class I suppressed profile, or with an RT profile, wherein the presence of the cells indicates that the tissue is injured.
  • this document features a nucleic acid array comprising at least 20 nucleic acid molecules, wherein each of the at least 20 nucleic acid molecules has a different nucleic acid sequence, and wherein at least 50 percent of the nucleic acid molecules of the array comprise a sequence from nucleic acid selected from the group consisting of the nucleic acids listed in Tables 1-14, 19, and 20.
  • the array can comprise at least 50 nucleic acid molecules, wherein each of the at least 50 nucleic acid molecules has a different nucleic acid sequence.
  • the array can comprise at least 100 nucleic acid molecules, wherein each of the at least 100 nucleic acid molecules has a different nucleic acid sequence.
  • Each of the nucleic acid molecules that comprise a sequence from nucleic acid selected from the group can comprise no more than three mismatches. At least 75 percent of the nucleic acid molecules of the array can comprise a sequence from nucleic acid selected from the group. At least 95 percent of the nucleic acid molecules of the array can comprise a sequence from nucleic acid selected from the group.
  • the array can comprise glass. The at least 20 nucleic acid molecules can comprise a sequence present in a human.
  • this document features a computer-readable storage medium having instructions stored thereon for causing a programmable processor to determine whether one or more nucleic acids listed in Tables 5-14, and the third column of Table 20 are present in a tissue sample at elevated levels.
  • the computer-readable storage medium can further comprise instructions stored thereon for causing a programmable processor to determine whether one or more of the nucleic acids listed in Tables 5-14, and the third column of 20 is expressed at a greater level in the tissue sample than in a control tissue sample.
  • this document features a computer-readable storage medium having instructions stored thereon for causing a programmable processor to determine whether one or more nucleic acids listed in Tables 1-4 and the third column of Table 19 are present in a tissue sample at decreased levels.
  • the computer-readable storage medium can further comprise instructions stored thereon for causing a programmable processor to determine whether one or more of the nucleic acids listed in Tables 1-4 and the third column of Table 19 is expressed at a lower level in the tissue sample than in a control tissue sample.
  • this document features an apparatus for determining whether a tissue is injured, the apparatus comprising: one or more collectors for obtaining signals representative of the presence of one or more nucleic acids listed in Tables 1-14, 19, and 20 in a sample from the tissue; and a processor for analyzing the signals and determining whether the tissue is injured.
  • the one or more collectors can be configured to obtain further signals representative of the presence of the one or more nucleic acids in a control sample.
  • this document features a method for detecting tissue rejection.
  • the method comprises, or consists essentially of, determining whether or not tissue transplanted into a mammal contains cells that express a reduced level of a cadherin polypeptide or a transporter polypeptide, wherein the presence of the cells indicates that the tissue is being rejected.
  • the mammal can be a human.
  • the tissue can be kidney tissue.
  • the tissue can be a kidney.
  • the method can comprise determining whether or not the tissue contains cells that express a reduced level of the cadherin polypeptide.
  • the cadherin polypeptide can be an E-cadherin polypeptide or a Ksp-cadherin polypeptide.
  • the method can comprise determining whether or not the tissue contains cells that express a reduced level of the transporter polypeptide.
  • the transporter polypeptide can be selected from the group consisting of Slc2a2, Slc2a4, Slc2a5 Slc5al, Slc5a2, Slc5alO, Slc7a7, Slc7a8, Slc7a9, Slc7alO, Slc7al2, Slc7al3, Slcla4, Slc3al, Slclal, aquaporin 1, aquaporin 2, aquaporin 3, aquaporin 4, ABC transporter (e.g., a member of the ABC transporter polypeptide family), solute carrier, and ATPase polypeptides.
  • ABC transporter e.g., a member of the ABC transporter polypeptide family
  • the determining step can comprise measuring the level of mRNA encoding the cadherin polypeptide or the transporter polypeptide.
  • the determining step can comprise measuring the level of the cadherin polypeptide or the transporter polypeptide.
  • the method can comprise determining whether or not the tissue contains cells that express the cadherin polypeptide or the transporter polypeptide at a level less than the average level of expression exhibited in cells from control tissue that has not been transplanted.
  • the determining step can comprise determining whether or not a sample contains the cells, wherein the sample comprises cells, was obtained from tissue that was transplanted into the mammal, and was obtained from the tissue within fifteen days of the tissue being transplanted into the mammal.
  • FIG 1 is a depiction of the algorithm used to develop the unique IRIT list.
  • FIG. 2 is a dendrogram for donor (implant) biopsies of 42 deceased donor (DD) and 45 living donor (LD) kidneys.
  • the DIANA dendrogram is based on all 7376 interquartile range- (IQR-) filtered probesets. Black boxes indicate pairs, and arrows indicate delayed graft function (DGF).
  • FIG 3 is a graph showing principal component analysis (PCA) of the transcriptome of 87 donor (implant) biopsies, based on the same set of 7376 IQR-f ⁇ ltered probesets as clustered in Figure 2.
  • PCA principal component analysis
  • FIG. 4 is a chart showing pathogenesis based transcript (PBT) scores calculated for the 3 clusters shown in Figure 2. Only those probesets passing the non-specific (IQR) filtering step were used to calculate the scores.
  • PBT scores are defined as fold-change relative to the nephrectomy controls, averaged over all probesets within each PBT.
  • FIG. 5 is a chart showing p-values from Bayesian t-tests comparing inter-cluster PBT scores, p-values have been corrected using Benjamini and Hochberg's false discovery rate method.
  • the Cluster 3 (“high-risk") group has been subdivided into samples with and without DGF.
  • FIG. 6 is a graph plotting ROC curves for Principal Component 1 (PCl), showing PCA 1 's value in predicting DGF status in the 42 DD kidneys.
  • PCl was based on all probesets passing the IQR-filter, and on all 87 (LD + DD) samples.
  • Solid line the smoothed-average ROC curve of all 42 leave-one-out cross validated (LOOCV) estimates; horizontal bars, medians in boxplots; dotted lines, non-smoothed individual ROC curves for each of the LOOCV estimates.
  • LOOCV smoothed-average ROC curve of all 42 leave-one-out cross validated
  • FIG. 7 is a graph plotting ROC curves showing individual PBT scores (RTs, tGRITs, and mCATs) and PCl scores in predicting DGF status in the 42 DD kidneys.
  • the PCl scores were based on genes that were both IQR filtered and PBTs. Horizontal bars, medians in boxplots; dotted lines, non-smoothed individual ROC curves for each of the LOOCV estimates.
  • FIG. 8. is a table showing the correlation of gene sets with function (GFR) at the time of biopsy and 3 months after biopsy.
  • FIG. 9. is a table showing the correlation of gene sets with the degree of loss of function/GFR before biopsy (all gene sets; center column) and recovery of function/GFR after biopsy (IRITs, GSTs, CISTs; right column).
  • FIG. 10 is a table showing that the best correlations between renal function (GFR) and gene sets are with the IRITs, particularly with IRITsD3 and IRITsD5.
  • FIG. 11 is a table showing that the best correlations between degree of loss of function/GFR and gene sets are with the IRITs, especially the IRITsD3 and IRITsD5.
  • FIG. 12 Histology of rejecting kidneys (CBA into B6 transplants; PAS staining).
  • F Day 21 transplant with marked tubulitis (arrows) and distorted tubules (magnification 10Ox).
  • FIG. 13 Real time RT-PCR analysis of CD 103 mRNA expression.
  • NCBA normal kidney
  • CBA into B6 normal kidney
  • NCBA contralateral CDlOS 7 TiOSt kidneys
  • Values are fold changes relative to control kidney (NCBA), expressed as mean ⁇ SE. Assays were done in duplicate.
  • FIG. 14 Histology of allografts rejecting in wild-type (CB A into Balb/c) or
  • CD103 7" (CBA into CD103 " ⁇ ) hosts at day 21 post transplant.
  • FIG. 15 Expression of epithelial transporter transcripts (glucose transporters, amino acid transporters, aquaporins) in isografts and rejecting allografts (CBA into B6) at days 5, 7, and 21 post-transplant, determined by Affymetrix microarrays MOE 430A.
  • FIG. 16 E-cadherin and Ksp-cadherin in rejecting allografts.
  • A) Real time RT- PCR analysis of mRNA expression of cadherins in rejecting kidney (CBA into B6). Values are fold changes relative to control (CBA) kidney, expressed as means ⁇ SE (n 2, three kidneys in each pool). Assays were done in duplicate.
  • B) Western blot analysis of E-cadherin and Ksp-cadherin expression. Fold changes were calculated from the band intensity ratio of Tx (transplant: CBA into B6) versus C (contralateral kidney: B6). Shown are means ⁇ SE, n 3.
  • C E-cadherin and Ksp-cadherin mRNA expression in allografts rejecting in wild-type Balb/c (WT) or CD1O3 "7" hosts at day 21 post transplant.
  • FIG. 17 Immunohistochemical staining of E-cadherin and Ksp-cadherin (magnification 10Ox). Arrows show localization of cadherins.
  • E-cadherin was localized to the basolateral membrane A) in B6 host kidney and B) in rejecting allografts (CBA into B6).
  • CBA into B6 B6 host kidney
  • CBA into B6 B6 host kidney
  • E-cadherin staining was decreased with some redistribution to the apical membrane C) in allografts rejecting in wild-type hosts (CBA into B6) and D) in allografts rejecting in CDl(B "7" hosts (CBA into CD103 "A ).
  • Ksp-cadherin was localized to the basolateral membrane in normal CBA kidney (control). Ksp-cadherin was decreased in rejecting allografts F) in wild-type hosts (CBA into B6) at day 7 post transplant, G) in wild-type hosts (CBA into B6) at day 21 post transplant and H) in CD 103 " ⁇ hosts (CBA into CD 103 " ⁇ ) at day 21 post transplant.
  • tissue injury e.g., injury inherent in a tissue to be transplanted, or tissue injury that may occur with organ transplantation, including alloimmune and non-alloimmune injury
  • this document provides methods and materials that can be used to determine whether a tissue is injured or susceptible to injury and delay in function.
  • a mammal can be diagnosed as having transplanted tissue that is injured (due to rejection or not) or likely to be injured if it is determined that the tissue contains cells that express altered levels of one or more nucleic acid transcripts, as described herein.
  • transcripts including mouse and human "injury-and-repair induced transcripts” (IRITs), “not in isografts injury-and-repair induced transcripts” (NIRITs), “gamma-interferon suppressed transcripts” (GSTs), and “class I suppressed transcripts” (CISTs) can be used to distinguish tissue (e.g., transplanted tissue) that is injured from tissue that is not injured.
  • IRITs injury-and-repair induced transcripts
  • NIRITs not in isografts injury-and-repair induced transcripts
  • GSTs gamma-interferon suppressed transcripts
  • CISTs class I suppressed transcripts
  • This document also is based, in part, on the discovery that the expression levels of mouse "cytotoxic T lymphocyte-associated transcripts" (CATs) and "true gamma-interferon dependent and rejection-induced transcripts” (tGRITs) can be used to distinguish tissue (e.g., transplanted tissue) that is being rejected from tissue that is not being rejected as disclosed, for example, in U.S. Publication Nos. 2006/0269948 and 2006/0269949.
  • the expression levels of nucleic acids listed in Tables 5-14 can be assessed in transplanted tissue to determine whether or not that transplanted tissue is injured.
  • RTs renal transcripts
  • Slcs solute carriers
  • a mammal can be diagnosed as having transplanted tissue that is injured if it is determined that the tissue contains cells expressing elevated levels of one or more IRITs, NIRITs, GSTs, and/or CISTs, or that express elevated levels one or more of the nucleic acids listed in Tables 5-14.
  • a mammal can be diagnosed as having transplanted tissue that is injured if it is determined that the tissue contains cells that express reduced levels of one or more Slcs and RTs listed in Tables 1-4.
  • a mammal can be diagnosed as having transplanted tissue that is injured if it is determined that the tissue contains cells that express gene lists and/or pathways that are significantly positively or negatively correlated with the gene profiles described in Tables 1-14 (e.g., the gene set in Table 19 and/or the gene pathways in Table 21 that correlate with the gene profile shown in Tables 1 and 2, or the gene set in Table 20 and/or the gene pathways in Table 22 that correlate with the gene profile in Tables 7 and 8).
  • the tissue contains cells that express gene lists and/or pathways that are significantly positively or negatively correlated with the gene profiles described in Tables 1-14 (e.g., the gene set in Table 19 and/or the gene pathways in Table 21 that correlate with the gene profile shown in Tables 1 and 2, or the gene set in Table 20 and/or the gene pathways in Table 22 that correlate with the gene profile in Tables 7 and 8).
  • IRITs injury and repair-induced transcripts
  • IRITs refers to transcripts that are increased in isografts at least once between day 1 and day 21, as compared to normal kidney, excluding allogeneic effects as well as T cell-associated, macrophage associated, and IFN- ⁇ inducible transcripts.
  • IRITs indicate non-alloimmune effects, such as injury caused by surgery or ischemia reperfusion, for example.
  • the ATN model discussed herein demonstrates ischemia reperfusion injury.
  • an "IRIT” is identified based on expression that is at least two-fold in kidney isografts as compared to normal kidney. Examples of IRITs include, without limitation, the nucleic acids listed in Tables 7-10. Some IRITs, such as those listed in Table 9, also are primary macrophage associated transcripts (MATs). These transcripts indicate non-alloimmune injury involving innate immune responses.
  • MATs primary macrophage associated transcripts
  • Some gene sets and pathways have been found to be positively or negatively correlated with IRITs.
  • the genes listed in the first column of Table 20 are negatively correlated with IRITs, while the genes listed in the third column of Table 20 are positively correlated with IRITs.
  • the pathways listed in the left column of Table 22 are negatively correlated with IRITs, while the pathways listed in the right column of Table 22 are positively correlated with IRITs.
  • increased expression of the positively correlated genes listed in Table 20 increased activity of the positively correlated pathways listed in Table 22
  • decreased expression of the negatively correlated genes listed in Table 20 or decreased activity of the negatively correlated pathways listed in Table 22 can indicate tissue injury (e.g., non-alloimmune injury).
  • NIRITs (not in isografts) injury and repair induced transcripts
  • NIRITs refers to transcripts that are elevated in kidney allografts vs. isografts at least once between day 1 and day 42 post transplant in WT hosts, excluding transcriptomes of infiltrating T cells, B cells and macrophages, IFN-K inducible genes, cytotoxic T cell associated transcripts, IFN- ⁇ dependent rejection induced transcripts, and transcripts showing strain differences.
  • a "NIRIT” can be identified based on expression that is increased in kidney allografts as compared to control kidneys, but not increased in kidney isografts as compared to control kidneys.
  • NIRITs indicate injury that occurs in the parenchyma of the kidney (i.e., the transcriptome of the infiltrating cell compartments have been "removed") and is due to an alloimmune response rather than a non- alloimmune response.
  • examples of NIRITs include, without limitation, the nucleic acids listed in Tables 5 and 6.
  • nucleic acids that are differentially expressed in tissue that is injured as compared to control tissue that is not injured can be nucleic acids that are suppressed by gamma interferon (IFN- ⁇ ).
  • IFN- ⁇ suppressed transcripts or "GSTs” as used herein refers to transcripts that are expressed in IFN- ⁇ receptor deficient kidney allograft tissue at a level that is greater than the level of expression in WT kidney allograft tissue.
  • a "GST” is identified based on expression that is increased at least two-fold in IFN- ⁇ receptor deficient kidney allograft tissue as compared to the level of expression in WT kidney allograft tissue.
  • GSTs indicate the underlying alternative inflammatory response to alloimmune and non-alloimmune injury. Examples of GSTs include, without limitation, the nucleic acids listed in Tables 11 and 12.
  • class-I proteins e.g., MHC class Ia and/or Ib proteins such as the Tapl transporter and beta 2 microglobulin.
  • class I suppressed transcripts or “CISTs” as used herein refers to transcripts that are expressed in class I protein (e.g., Tapl transporter and beta 2 microglobulin) deficient kidney allograft tissue at a level that is greater than the expression in WT kidney allograft tissue.
  • a "CIST" is identified based on expression that is increased at least two-fold in class I deficient kidney allograft tissue as compared to the level of expression in WT kidney allograft tissue.
  • CISTs indicate the underlying alternative inflammatory response that occurs to alloimmune and non-alloimmune injury, and demonstrates the involvement of IFN-K in the process.
  • Examples of CISTs include, without limitation, the nucleic acids listed in Tables 13 and 14.
  • a nucleic acid can be included in two or more of the categories described herein. For example, some nucleic acids can be considered to be
  • GSTs and CISTs Elevated levels of such GST/CIST nucleic acids can indicate injury in allograft transplants, for example.
  • the RTs listed in Tables 3 and 4 are renal transcripts that are reduced in allografts and isografts with injury. These transcripts reflect non-alloimmune injury due, for example, to surgical stress, ischemia reperfusion, and other causes, as well as ongoing additional injury effects that occur in alloimmune rejection.
  • the Slcs listed in Tables 1 and 2 are renal solute carrier transcripts that are decreased in allografts and isografts with injury. Like the RTs, the Slcs reflect non-alloimmune injury and alloimmune injury. Some gene sets and pathways have been found to be positively or negatively correlated with Slcs.
  • the genes listed in the first column of Table 19 are negatively correlated with Slcs, while the genes listed in the third column of Table 19 are positively correlated with Slcs.
  • the pathways listed in the left column of Table 21 are negatively correlated with Slcs, while the pathways listed in the right column of Table 21 are positively correlated with Slcs.
  • reduced expression of the positively correlated genes listed in Table 19 reduced activity of the positively correlated pathways listed in Table 21, increased expression of the negatively correlated genes listed in Table 19, or increased activity of the negatively correlated pathways listed in Table 21 can indicate tissue injury (e.g., non-alloimmune injury or alloimmune injury).
  • cytotoxic T lymphocyte-associated transcripts or "CATs” refers to transcripts that are not usually expressed in kidney but are induced in rejection, and that may reflect T cells recruited to the graft.
  • Examples of CATs include, without limitation, the nucleic acids listed in Table 15. These transcripts are diagnostic for allograft rejection and are referred to in co- pending U.S. Publication No. 2006/0269948.
  • nucleic acids can be regulated by IFN- ⁇ and induced by rejection.
  • the term "true interferon gamma dependent and rejection-induced transcripts” or “tGRITs” refers to rejection-induced transcripts that are IFN- ⁇ -dependent in rejection, and also are unique transcripts that are increased at least 2-fold by rIFN- ⁇ . See, co-pending U.S. Publication No. 2006/0269949. Examples of tGRITs include, without limitation, the nucleic acids listed in Table 16, which can be diagnostic for allograft rejection.
  • the term "transcript” as used herein refers to an mRNA identified by one or more numbered Affymetrix probe sets, while a "unique transcript” is an mRNA identified by only one probe set.
  • a tissue can be identified as being injured if it is determined that the tissue contains cells having an injury and repair profile, a not-in- isografts injury and repair profile, an IFN-K suppressed profile, or a class I suppressed profile.
  • the term "injury and repair profile” refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 7-10 is present at an elevated level.
  • a sample e.g., a sample of tissue that is transplanted or is to be transplanted
  • one or more than one e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more
  • not-in-isografts injury and repair profile refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 5 and 6 is present at an elevated level.
  • a sample e.g., a sample of tissue that is transplanted or is to be transplanted
  • one or more than one e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more
  • IFN-K suppressed profile refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 11 and 12 is present at an elevated level.
  • a sample e.g., a sample of tissue that is transplanted or is to be transplanted
  • one or more than one e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more
  • class I suppressed profile refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 13 and 14 is present at an elevated level.
  • a sample e.g., a sample of tissue that is transplanted or is to be transplanted
  • one or more than one e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more
  • a tissue can be identified as being injured if it is determined that the tissue contains cells having a RT profile or a SIc profile.
  • RT profile refers to a nucleic acid or polypeptide profile in a sample
  • SIc profile refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 3 and 4 is present at a reduced level
  • SIc profile refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 1 and 2 is present at an reduced level.
  • a tissue can be identified as being injured if it is determined that the tissue contains cells having a quantitative injury and repair profile, a quantitative not-in-isografts injury and repair profile, a quantitative IFN-K suppressed profile, or a quantitative class I suppressed profile.
  • quantitative injury and repair profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 7-10 are present at an elevated level.
  • a quantitative human injury and repair profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 8 are present at an elevated level.
  • Quantitative not-in-isografts injury and repair profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 5 and 6 are present at an elevated level.
  • a human not-in-isografts injury and repair profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 6 are present at an elevated level.
  • quantitative IFN-K suppressed profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 11 and 12 are present at an elevated level.
  • a human IFN-K suppressed profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 12 are present at an elevated level.
  • quantitative class I suppressed profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 13 and 14 are present at an elevated level.
  • a human class I suppressed profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 14 are present at an elevated level.
  • a tissue can be identified as being injured if it is determined that the tissue contains cells having a quantitative RT profile, or a quantitative SIc profile.
  • quantitative RT profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 3 and 4 are present at a reduced level.
  • a quantitative human RT profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 4 are present at a reduced level.
  • Quantitative SIc profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 1 and 2 are present at a reduced level.
  • a quantitative human SIc profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 2 are present at a reduced level.
  • a tissue can be identified as being injured if it is determined that the tissue contains cells having an injury and repair positively correlated profile or an injury and repair negatively correlated profile.
  • the term "injury and repair positively correlated profile” refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) of the nucleic acids or polypeptides encoded by the nucleic acids listed in column 3 of Table 20 is present at an elevated level.
  • a sample e.g., a sample of tissue that is transplanted or is to be transplanted
  • one or more than one e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25
  • the term "injury and repair negatively correlated profile” refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) of the nucleic acids or polypeptides encoded by the nucleic acids listed in column 1 of Table 20 is present at an elevated level.
  • the presence of an injury and repair positively correlated profile can indicate that a tissue is injured.
  • the presence of an injury and repair negatively correlated profile also can indicate that a tissue is injured.
  • a tissue can be identified as being injured if it is determined that the tissue contains cells having a quantitative injury and repair positively correlated profile or a quantitative injury and repair negatively correlated profile.
  • quantitative injury and repair positively correlated profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 20 are present at an elevated level.
  • a quantitative injury and repair positively correlated profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 20 are present at an elevated level.
  • quantitative injury and repair negatively correlated profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 20 are present at an elevated level.
  • a quantitative injury and repair negatively correlated profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 20 are present at an elevated level.
  • a tissue can be identified as being injured if it is determined that the tissue contains cells having an SIc positively correlated profile or an SIc negatively correlated profile.
  • SIc positively correlated profile refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 19 is present at a reduced level.
  • a sample e.g., a sample of tissue that is transplanted or is to be transplanted
  • one or more than one e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25
  • SIc negatively correlated profile refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 19 is present at a reduced level.
  • the presence of an SIc positively correlated profile can indicate that a tissue is injured.
  • the presence of an SIc negatively correlated profile also can indicate that a tissue is injured.
  • a tissue can be identified as being injured if it is determined that the tissue contains cells having a quantitative SIc positively correlated profile or a quantitative SIc negatively correlated profile.
  • quantitative SIc positively correlated profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 19 are present at a reduced level.
  • a quantitative SIc positively correlated profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 19 are present at a reduced level.
  • quantitative SIc negatively correlated profile refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 19 are present at a reduced level.
  • a quantitative SIc negatively correlated profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 19 are present at a reduced level.
  • tissue injury e.g., tissue rejection
  • any mammal including, without limitation, a human, monkey, horse, dog, cat, cow, pig, mouse, or rat.
  • the methods and materials provided herein can be used to detect injury of any type of tissue including, without limitation, kidney, heart, liver, pancreas, and lung tissue.
  • the methods and materials provided herein can be used to determine whether or not a human who received a kidney transplant is experiencing injury of the transplanted kidney.
  • sample containing cells can be used to determine whether or not transplanted tissue, tissue that is not transplanted, or tissue that is to be transplanted (e.g., donor biopsy) contains cells that express one or more IRITs, NIRITs, GSTs, and or CISTs, or that express one or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 5-14, at elevated levels.
  • any type of sample containing cells can be used to determine whether or not transplanted tissue, tissue that is not transplanted, or tissue that is to be transplanted (e.g., donor biopsy) contains cells that express one or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 1-4 at decreased levels.
  • any type of sample containing cells can be used to determine whether transplanted tissue, tissue that is not transplanted, or tissue that is to be transplanted (e.g., donor biopsy) contains cells that express one or more nucleic acids that significantly positively or negatively correlate with nucleic acids listed in Tables 1-14.
  • biopsy e.g., punch biopsy, aspiration biopsy, excision biopsy, needle biopsy, or shave biopsy
  • tissue section e.g., aspiration biopsy, excision biopsy, needle biopsy, or shave biopsy
  • tissue section e.g., lymph fluid samples
  • a lymph fluid sample can be obtained from one or more lymph vessels that drain from the tissue.
  • a sample can contain any type of cell including, without limitation, cytotoxic T lymphocytes, CD4 + T cells, B cells, peripheral blood mononuclear cells, macrophages, kidney cells, lymph node cells, or endothelial cells. Additional examples of Slcs, RTs, IRITs, NIRITs, GSTs, and CISTs, as well as other transcripts with altered expression levels in injured tissues (e.g., genes in pathways related to glutathione metabolism, fatty acid elongation, and cell communication) can be identified using the procedures described herein.
  • the procedures described in Examples 1 and 2 can be used to identify RTs other than those listed in Tables 1 -4
  • the procedures described in Examples 1 and 4 can be used to identify IRITs other than those listed in Tables 7-10
  • the procedures described in Examples 1 and 3 can be used to identify NIRITs other than those listed in Tables 5 and 6
  • the procedures described in Examples 1 and 5 can be used to identify GSTs other than those listed in Tables 11 and 12
  • the procedures described in Examples 1 and 6 can be used to identify CISTs other than those listed in Tables 13 and 14.
  • any number of Slcs, RTs, IRITs, NIRITs, GSTs, CISTs, or nucleic acids listed in Tables 1-14, 19, 20, 21, and 22 can be evaluated to determine whether or not transplanted tissue is injured.
  • the expression of one or more than one (e.g., two, three, four, five, six, seven, eight, nine, ten, 15, 20, 25, 30, 40, 50, 75, 100, or more than 100) of the nucleic acids listed in Tables 1-14, 19, 20, 21, and 22 can be used.
  • an elevated level of a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 5-14 is any level that is greater than a reference level for that nucleic acid or polypeptide.
  • an elevated level of a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 5-14 can be about 0.3, 0.5, 0.7, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.2, 2.4, 2.6, 2.8, 3, 3.3, 3.6, 4, 4.5, 5, 5.5, 6, 7, 8, 9, 10, 15, 20, or more times greater than the reference level for that nucleic acid or polypeptide, respectively.
  • reduced level as used herein with respect to the level of a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 1-4 is any level that is less than a reference level for that nucleic acid or polypeptide.
  • a reduced level of a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 1-4 can be about 0.3, 0.5, 0.7, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.2, 2.4, 2.6, 2.8, 3, 3.3, 3.6, 4, 4.5, 5, 5.5, 6, 7, 8, 9, 10, 15, 20, or more times less than the reference level for that nucleic acid or polypeptide, respectively.
  • the term "reference level" as used herein with respect to a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 1-14 is the level of that nucleic acid or polypeptide typically expressed by cells in tissues that are free of injury.
  • a reference level of a nucleic acid or polypeptide can be the average expression level of that nucleic acid or polypeptide, respectively, in cells isolated from kidney tissue that has not been injured.
  • a reference level can be any amount.
  • a reference level can be zero. In this case, any level greater than zero would be an elevated level.
  • samples can be used to determine a reference level.
  • cells obtained from one or more healthy mammals e.g., at least 5, 10, 15, 25, 50, 75, 100, or more healthy mammals
  • levels from comparable samples are used when determining whether or not a particular level is an elevated or reduced level.
  • levels from one type of cells are compared to reference levels from the same type of cells.
  • levels measured by comparable techniques are used when determining whether or not a particular level is an elevated level or a reduced level.
  • any suitable method can be used to determine whether or not a particular nucleic acid is expressed at a detectable level or at a level that is greater or less than the average level of expression observed in control cells.
  • expression of a particular nucleic acid can be measured by assessing mRNA expression.
  • mRNA expression can be evaluated using, for example, northern blotting, slot blotting, quantitative reverse transcriptase polymerase chain reaction (RT-PCR), real-time RT-PCR, or chip hybridization techniques.
  • Methods for chip hybridization assays include, without limitation, those described herein. Such methods can be used to determine simultaneously the relative expression levels of multiple mRNAs.
  • expression of a particular nucleic acid can be measured by assessing polypeptide levels.
  • polypeptide levels can be measured using any method such as immuno- based assays (e.g., ELISA), western blotting, or silver staining.
  • a sample obtained from a donor at any time prior to transplantation can be assessed for the presence of cells expressing elevated levels of a nucleic acid listed in Tables 5-14, decreased levels of a nucleic acid listed in Tables 1-4, or significant alterations in gene profiles that correlate with genes listed Tables 1-14 (such as the gene profiles and pathways referred to in Tables 19, 20, 21, and 22).
  • a sample can be obtained from a donor 1, 2, 3, 4, 5, 6, 7, or more than 7 days prior to transplant, or can be obtained from a donor tissue within hours (e.g., 1, 2, 3, 4, 6, 8, or 12 hours) prior to transplantation.
  • a sample obtained from transplanted tissue at any time following the tissue transplantation can be assessed for the presence of cells expressing elevated levels of a nucleic acid listed in Tables 5-14, or decreased levels of a nucleic acid listed in Tables 1-4.
  • a sample can be obtained from transplanted tissue 1, 2, 3, 4, 5, 6, 7, 8, or more hours after the transplanted tissue was transplanted, hi some cases, a sample can be obtained from transplanted tissue one or more days (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 42, or more days) after the transplanted tissue was transplanted.
  • transplanted tissue 1, 2, 3, 4, 5, 6, 7, 8, or more hours after the transplanted tissue was transplanted, hi some cases, a sample can be obtained from transplanted tissue one or more days (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 42, or more days) after the transplanted tissue was transplanted.
  • a sample can be obtained from transplanted tissue 1 to 7 days (e.g., 1 to 3 days, or 5 to 7 days) after transplantation and assessed for the presence of cells expressing elevated levels of one or more IRITs, NIRITs, GSTs, or CISTs, expressing elevated levels of one or more nucleic acids listed in Tables 5-14, expressing decreased levels of one or more transcripts listed in Tables 1-4, or expressing significant alterations in gene profiles that correlate with genes listed Tables 1-14 (such as those gene profiles and/or pathways referred to in Tables 19, 20, 21, and 22).
  • a mammal can be diagnosed as having transplanted tissue that is being rejected if it is determined that the mammal or tissue contains cells that express a reduced level of a cadherin polypeptide or a transporter polypeptide.
  • Any type of sample containing cells can be used to determine whether or not the mammal or transplanted tissue contains cells that express a reduced level of a cadherin polypeptide or a transporter polypeptide.
  • biopsy e.g., punch biopsy, aspiration biopsy, excision biopsy, needle biopsy, or shave biopsy
  • tissue section e.g., lymph fluid, blood, and synovial fluid samples
  • a tissue biopsy sample can be obtained directly from the transplanted tissue.
  • a lymph fluid sample can be obtained from one or more lymph vessels that drain from the transplanted tissue.
  • a sample can contain any type of cell including, without limitation, cytotoxic T lymphocytes, CD4 + T cells, B cells, peripheral blood mononuclear cells, macrophages, kidney cells, lymph node cells, or endothelial cells.
  • cadherin polypeptides include, without limitation, E-cadherin polypeptides, Ksp-cadherin polypeptides, and any other cadherin polypeptide.
  • transporter polypeptides include, without limitation, Slc2a2, Slc2a4, Slc2a5 Slc5al, Slc5a2, Slc5alO, Slc7a7, Slc7a8, Slc7a9, Slc7alO, Slc7al2, Slc7al3, Slcla4, Slc3al, Slclal, aquaporins (e.g., aquaporin 1, aquaporin 2, aquaporin 3, and aquaporin 4), members of the family of ABC transporters, solute carriers, and ATPases.
  • aquaporins e.g., aquaporin 1, aquaporin 2, aquaporin 3, and aquaporin 4
  • any number of polypeptides disclosed herein or nucleic acids encoding such polypeptides can be evaluated to determine whether or not transplanted tissue will be rejected.
  • the expression of one or more than one (e.g., two, three, four, five, six, seven, eight, nine, ten, 15, 20, 25, 30, 40, 50, 75, 100, or more than 100) of the transporter polypeptides provided herein can be used.
  • determining that a polypeptide is expressed at a reduced level in a sample can indicate that transplanted tissue will be rejected.
  • transplanted tissue can be evaluated by determining whether or not the tissue contains cells that express one or more cadherin or transporter polypeptides at a level that is less than the average expression level observed in control cells obtained from tissue that has not been transplanted.
  • a polypeptide can be classified as being expressed at a level that is less than the average level observed in control cells if the expression levels differ by at least 1-fold (e.g., 1.5-fold, 2-fold, 3-fold, or more than 3-fold).
  • Control cells typically are the same type of cells as those being evaluated.
  • the control cells can be isolated from kidney tissue that has not been transplanted into a mammal. Any number of tissues can be used to obtain control cells.
  • control cells can be obtained from one or more tissue samples (e.g., at least 5, 6, 7, 8, 9, 10, or more tissue samples) obtained from one or more healthy mammals (e.g., at least 5, 6, 7, 8, 9, 10, or more healthy mammals).
  • any appropriate method can be used to determine whether or not a particular polypeptide is expressed at a reduced level as compared to the average level of expression observed in control cells.
  • expression of a particular polypeptide can be measured by assessing mRNA expression.
  • mRNA expression can be evaluated using, for example, northern blotting, slot blotting, quantitative reverse transcriptase polymerase chain reaction (RT-PCR), real-time RT-PCR, or microarray chip hybridization techniques.
  • Methods for microarray chip hybridization assays include, without limitation, those described herein. Such methods can be used to determine simultaneously the relative expression levels of multiple mRNAs.
  • expression of a particular polypeptide can be measured by assessing polypeptide levels.
  • polypeptide levels can be measured using any method such as immuno- based assays (e.g., ELISA and immunohistochemistry), western blotting, or silver staining.
  • a sample obtained from transplanted tissue at any time following the tissue transplantation can be assessed for the presence of cells expressing a reduced level of a polypeptide provided herein, hi some cases, a sample can be obtained from transplanted tissue 1, 2, 3, 4, 5, 6, 7, 8, or more hours after the transplanted tissue was transplanted. In some cases, a sample can be obtained from transplanted tissue one or more days (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, or more days) after the transplanted tissue was transplanted.
  • a sample can be obtained from transplanted tissue 2 to 7 days (e.g., 5 to 7 days) after transplantation and assessed for the presence of cells expressing a reduced level of a polypeptide provided herein.
  • a biopsy can be obtained any time after transplantation if a patient experiences reduced graft function.
  • Ksp-cadherin mRNA and protein were decreased early, before the onset of tubulitis, coincident with interstitial infiltration. These results demonstrate that the decrease in Ksp-cadherin and E-cadherin can be attributed to the response of the epithelium to the inflammatory processes, responses that can permit the entry of inflammatory cells into the epithelium, and if unchecked can culminate in EMT.
  • T cell-mediated rejection in the interstitium can induce expression of effectors (e.g., TGF- ⁇ l, actins, vimentin, MMP2, collagens, hyaluronic acid, and many others) that can cause the tubule epithelium to change, permitting the interstitial inflammatory cells to enter the epithelium.
  • effectors e.g., TGF- ⁇ l, actins, vimentin, MMP2, collagens, hyaluronic acid, and many others
  • the effector T cell/macrophage infiltrate can deliver this contact-independent signal to the epithelium via soluble factors or via matrix- or even microcirculation changes.
  • the mechanism by which the interstitial CTL trigger epithelial changes can be that Tgfbl plays a role.
  • Tgfbl is produced by CTL and is expressed in a CTL line and in recently generated allogeneic cultures, and potentially by macrophages and by many cells in the graft.
  • the early increase in Tgfbl in isografts can exaggerate in allografts, and some Tgfbl -inducible transcripts can be greatly increased in rejecting allografts.
  • TGF- ⁇ l can trigger a decrease in cadherin expression and alterations in epithelial function.
  • the arrays provided herein can be two-dimensional arrays, and can contain at least 10 different nucleic acid molecules (e.g., at least 20, at least 30, at least 50, at least 100, or at least 200 different nucleic acid molecules).
  • Each nucleic acid molecule can have any length.
  • each nucleic acid molecule can be between 10 and 250 nucleotides (e.g., between 12 and 200, 14 and 175, 15 and 150, 16 and 125, 18 and 100, 20 and 75, or 25 and 50 nucleotides) in length.
  • each nucleic acid molecule can have any sequence.
  • the nucleic acid molecules of the arrays provided herein can contain sequences that are present within the nucleic acids listed in Tables 1-14, 19, and 20.
  • a sequence is considered present within a nucleic acid listed in, for example, Table 1 when the sequence is present within either the coding or non-coding strand.
  • both sense and anti-sense oligonucleotides designed to human Slc39a5 nucleic acid are considered present within Scl39a5 nucleic acid.
  • At least 25% (e.g., at least 30%, at least 40%, at least 50%, at least 60%, at least 75%, at least 80%, at least 90%, at least 95%, or 100%) of the nucleic acid molecules of an array provided herein contain a sequence that is (1) at least 10 nucleotides (e.g., at least 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, or more nucleotides) in length and (2) at least about 95 percent (e.g., at least about 96, 97, 98, 99, or 100) percent identical, over that length, to a sequence present within a nucleic acid listed in any of Tables 1-16.
  • an array can contain 100 nucleic acid molecules located in known positions, where each of the 100 nucleic acid molecules is 100 nucleotides in length while containing a sequence that is (1) 30 nucleotides in length, and (2) 100 percent identical, over that 30 nucleotide length, to a sequence of one of the nucleic acids listed in any of Tables 1-14, 19, and 20.
  • a nucleic acid molecule of an array provided herein can contain a sequence present within a nucleic acid listed in any of Tables 1-14, 19, and 20, where that sequence contains one or more (e.g., one, two, three, four, or more) mismatches.
  • the nucleic acid arrays provided herein can contain nucleic acid molecules attached to any suitable surface (e.g., plastic or glass), hi addition, any method can be use to make a nucleic acid array. For example, spotting techniques and in situ synthesis techniques can be used to make nucleic acid arrays. Further, the methods disclosed in U.S. Patent Nos. 5,744,305 and 5,143,854 can be used to make nucleic acid arrays. This description also provides methods and materials involved in determining the potential for recovery of organ function following injury. For example, Figure 8 shows that the SIc, RT, IRIT, GST and CIST gene sets correlate with function (glomerular filtration rate; GFR) at the time of biopsy and at 3 months after the biopsy.
  • GFR glomerular filtration rate
  • Figure 9 shows that gene sets correlate with the degree of loss of function/GFR before the biopsy (SLCs, RT's, IRITs, ST's, CISTs), as well as with recovery of function/GFR after the biopsy (IRITs, GSTs, CISTs).
  • Figures 10 and 11 show that the best correlation between renal function and gene sets are with the IRITs, especially with IRITsD3 and IRITsD5 (refer to Table 7 (mouse) and Table 8 (human)).
  • This document also provides methods and materials to assist medical or research professionals in determining whether or not a tissue is injured, is at increased risk for developing DGF following transplantation, or is likely to recover from alloimmune or non-alloimmune injury.
  • Medical professionals can be, for example, doctors, nurses, medical laboratory technologists, and pharmacists.
  • Research professionals can be, for example, principle investigators, research technicians, postdoctoral trainees, and graduate students.
  • a professional can be assisted by (1) determining the level of one or more nucleic acids or polypeptides encoded by nucleic acids listed in Tables 1-14, determining the level of a cadherin polypeptide, or determining the level of a transporter polypeptide in a sample, and (2) communicating information about that level to that professional.
  • Any method can be used to communicate information to another person (e.g., a professional).
  • information can be given directly or indirectly to a professional.
  • any type of communication can be used to communicate the information.
  • mail, e-mail, telephone, and face-to-face interactions can be used.
  • the information also can be communicated to a professional by making that information electronically available to the professional.
  • the information can be communicated to a professional by placing the information on a computer database such that the professional can access the information.
  • the information can be communicated to a hospital, clinic, or research facility serving as an agent for the professional.
  • Computer-readable medium and an apparatus for predicting rejection This disclosure further provides a computer-readable storage medium configured with instructions for causing a programmable processor to determine whether a tissue that has been or is to be transplanted is injured, and/or to determine the potential for recovery of organ function.
  • the determination of whether a tissue is injured can be carried out as described herein; that is, by determining whether one or more of the nucleic acids listed in Tables 5-14 and the third column of Table 20 is detected in a sample (e.g., a sample of the tissue), or expressed at a level that is greater than the level of expression in a corresponding control tissue, or by determining whether one or more of the nucleic acids listed in Tables 1-4 and the third column of Table 19 is expressed at a level that is less than the level of expression in a corresponding control tissue, hi some cases, it can be determined whether a tissue is being rejected by determining whether or not the tissue contains cells that express a reduced level of a cadherin polypeptide or a transporter polypeptide.
  • the processor also can be designed to perform functions such as removing baseline noise from detection signals.
  • Instructions carried on a computer-readable storage medium can be implemented in a high level procedural or object oriented programming language to communicate with a computer system. Alternatively, such instructions can be implemented in assembly or machine language. The language further can be compiled or interpreted language.
  • the nucleic acid detection signals can be obtained using an apparatus (e.g., a chip reader) and a determination of tissue injury can be generated using a separate processor (e.g., a computer).
  • a separate apparatus having a programmable processor can both obtain the detection signals and process the signals to generate a determination of whether injury is occurring or is likely to occur, hi addition, the processing step can be performed simultaneously with the step of collecting the detection signals (e.g., "realtime"). Any suitable process can be used to determine whether a tissue that has been or is to be transplanted is injured.
  • a process can include determining whether a pre-determined number (e.g., one, two, three, four, five, six, seven, eight, nine, ten, 15, 20, 25, 30, 40, 50, 75, 100, or more than 100) of the nucleic acids listed in Tables 5-14 and the third column of Table 20 is expressed in a sample (e.g., a sample of transplanted tissue) at a level that is greater than the average level observed in control cells (e.g., cells obtained from tissue that has not been transplanted or is not to be transplanted, or in a control transplanted tissue).
  • a pre-determined number e.g., one, two, three, four, five, six, seven, eight, nine, ten, 15, 20, 25, 30, 40, 50, 75, 100, or more than 100
  • the tissue can be determined to be injured and the potential for recovery of organ/tissue function can be determined to be low, depending on the gene sets that are predominantly altered. If the number of nucleic acids that are expressed in the sample is less than the pre-determined number, the tissue can be determined not to be injured.
  • the steps of this process e.g., the detection, or non-detection, of each of the nucleic acids
  • An apparatus for determining whether a tissue that has been or is to be transplanted can include, for example, one or more collectors for obtaining signals from a sample (e.g., a sample of nucleic acids hybridized to nucleic acid probes on a substrate such as a chip) and a processor for analyzing the signals and determining whether rejection will occur.
  • the collectors can include collection optics for collecting signals (e.g., fluorescence) emitted from the surface of the substrate, separation optics for separating the signal from background focusing the signal, and a recorder responsive to the signal, for recording the amount of signal.
  • the collector can obtain signals representative of the presence of one or more nucleic acids listed in Tables 1-14, 19, and 20 (e.g., in samples from transplanted and/or non-transplanted tissue).
  • the apparatus further can generate a visual or graphical display of the signals, such as a digitized representation.
  • the apparatus further can include a display. In some embodiments, the apparatus can be portable.
  • mice kidney allograft model that develops pathologic lesions that are diagnostic in human graft rejection.
  • a comparison of mouse kidney pathology to the mouse transcriptome was used to guide understanding of the relationship of lesions to transcriptome changes in human rejection.
  • mice Male CBA/J (CBA) and C57B1/6 (B6) mice were obtained from the Jackson Laboratory (Bar Harbor, ME). IFN- ⁇ deficient mice (BALB/c.GKO) and (B6.129S7-IFN ⁇ tmlTs ; B6.GKO) were bred in the Health Sciences Laboratory Animal Services at the University of Alberta. Mouse maintenance and experiments were in conformity with approved animal care protocols. CBA (H-2K, I-A k ) into C57B1/6 (B6; H- 2K b D b , I-A b ) mice strain combinations, BALB/c.GKO into B6.GKO were studied across full MHC and non-MHC disparities.
  • Renal transplantation was performed as a non life- supporting transplant model. Recovered mice were killed at day 1, 2, 3, 4, 5, 7, 14, 21 or 42 post-transplant. Kidneys were removed, snap frozen in liquid nitrogen and stored at -70°C. No mice received immunosuppressive therapy. Kidneys with technical complications or infection at the time of harvesting were removed from the study.
  • ATN Acute Tubular Necrosis
  • kidneys showed severe acute tubular injury with flattening of tubular epithelium, variation in cell size and shape, cellular swelling, loss of PAS positive brush borders, and individual tubular epithelial cell necrosis with denudation of the epithelium from the basement membrane and shedding of granular cellular debris into the tubular lumen.
  • tubular regenerative changes with nuclear enlargement, prominent nucleoli, and mitotic figures were observed.
  • Kidneys with ATN also showed interstitial edema and a focal minimal interstitial mononuclear cell infiltrate.
  • Microarrays High-density oligonucleotide GeneChip 430A and 430 2.0 arrays, GeneChip T7-Oligo(dT) Promoter Primer Kit, Enzo BioArray HighYield RNA
  • Transcript Labeling Kit IVT Labeling KIT, GeneChip Sample Cleanup Module, IVT cRNA Cleanup Kit were purchased from Affymetrix (Santa Clara, CA).
  • RNeasy Mini Kit was from Qiagen (Valencia, CA), Superscript II, E. coli DNA ligase, E. coli DNA polymerase I, E. coli RNase H, T4 DNA polymerase, 5X second strand buffer, and dNTPs were from Invitrogen Life Technologies.
  • RNA preparation and hybridization Total RNA was extracted from individual kidneys using the guanidinium-cesium chloride method and purified RNA using the RNeasy Mini Kit (Qiagen). RNA yields were measured by UV absorbance. The quality was assessed by calculating the absorbance ratio at 260 nm and 280 nm, as well as by using an Agilent Bio Analyzer to evaluate 18 S and 28S RNA integrity.
  • RNA from 3 mice was pooled.
  • RNA processing, labeling and hybridization to MOE430 2.0 arrays was carried out according to the protocols included in the Affymetrix GeneChip Expression Analysis Technical Manual (available on the World Wide Web at affymetrix.com).
  • cRNA used for Moe 430 2.0 arrays was labeled and fragmented using an IVT Labeling Kit and IVT cRNA Cleanup Kit.
  • Example 2 Renal transcripts (RTs) and Solute Carriers (Slcs)
  • RTs Renal transcripts
  • Slcs Solute Carriers
  • the changes in epithelial morphology likely reflect the effects of the T cell mediated interstitial inflammatory reaction, analogous to delayed type hypersensitivity (DTH).
  • DTH delayed type hypersensitivity
  • Morphologic lesions tubulitis, tubular shrinkage, loss of cadherins, and loss of polarity
  • Microarrays were used to explore the early transcriptome changes of renal parenchymal cells in mouse allografts and isografts, their relationship to the evolution of histologic lesions such as tubulitis, and their relationship to immunologic effector mechanisms.
  • To analyze expression of transcripts that reflect changes in the epithelium two sets of transcripts with high expression in normal kidney and low expression in inflammatory cells were selected. As a first set, epithelial transporters were selected because of their well documented importance for renal function. In particular, studies were focused specifically on the family of Slcs because of their extensive annotation.
  • T cell infiltrate in allografts was detectable from day 1 , and extended to the interstitium from days 5 to 7 post transplant, but morphologic epithelial changes did not develop until day 7. Transcripts for most Slcs were reduced in both allografts and isografts in response to transplant injury, but the loss was more severe and progressive in allografts and paralleled the development of tubulitis and other histologic lesions in the epithelium.
  • Mouse Slcs are listed in Table 1 ; humanized versions of the mouse Slcs are listed in Table 2.
  • Weighted sum decomposition of the SIc transcript set identified allospecific changes from day 1 and revealed multiple components of the allospecific epithelial response: sustained and progressive loss of transcripts, and lack of a positive response to injury.
  • SIc subsets with specific biological functions transporters of glucose, amino acids, organic ions, metal ions, Na, NaHCl, monocarboxyl acids, and mitochondrial transporters
  • All subgroups showed a strikingly similar expression pattern in both isografts and allografts, respectively, resembling the pattern with loss of transcripts described earlier for the entire SIc set.
  • RTs renal transcripts
  • Loss of transcripts was not attributable to simple dilution and affected the majority of renal transcripts, representing a selective structured program that leads to loss of at least some products and presumably function.
  • the early changes in the transcriptome of renal parenchymal cells reflect the same mechanisms as the later development of histologic lesions such as tubulitis: loss of renal transcripts was dependent on the alloimmune response and T cells, but independent of IFN-K, Prfl , GzmA, GzmB, and alloantibody.
  • the loss of epithelial transcripts should offer a system for objectively measuring the changes in renal allograft biopsies that can add to the current Banff system of grading morphologic lesions.
  • genes during the alloresponse alone were investigated, excluding transcriptomes of infiltrating T cells, B cells and macrophages. Genes inducible by IFN- ⁇ and genes activated in the isografts also were excluded.
  • all transcripts increased in at least one of the allograft conditions i.e., day 1, 2, 3, 4, 5, 7, 14, 21, or 42 post transplant.
  • This list then was corrected for IRIT (injury and repair induced transcripts - induced in the isografts), CAT (cytotoxic T cell associated transcripts), GRIT (gamma interferon dependent rejection induced transcripts), MAT (macrophage associated transcripts), BAT (B cell associated transcripts including immunoglobulin transcripts), and transcripts showing strain differences, using all probe sets corresponding to genes present in these lists.
  • the final NIRIT list included 714 nonredundant genes (Table 5 lists the mouse genes; Table 6 lists the humanized versions of the mouse genes).
  • IRIT Injury and Repair Induced Transcripts
  • IRITs showed enrichment in GeneSpring Gene Ontology (GO) categories related to morphogenesis, extracellular matrix, response to stress and cell cycle.
  • the expression pattern of IRITs showed significant correlations with the KEGG pathways, including TGF ⁇ signaling, apoptosis, and cell cycle.
  • IRIT-Dl The time course of IRIT expression was de-convoluted into three profiles, designated IRIT-Dl , IRIT-D3 and IRIT-D5, which were characterized by peak expression in particular days post-transplant (refer to Table 7).
  • the IRIT-Dl profile showed enrichment in systemic response and epithelium development
  • IRIT- D3 showed enrichment in stress response, epithelium development, and mesenchyme differentiation
  • IRIT-D 5 represented stress response, extracellular matrix, cell cycle, TGF ⁇ signaling, epithelial development, and mesenchyme differentiation.
  • MATs primary macrophages associated transcripts
  • GCOS GCOS method
  • Transcripts were required to be flagged as present, increased > 5-fold over the NB6 kidneys in at least one of the culture conditions, and have ae raw signal in NB6 and NCBA kidney below 200.
  • the resulting list contained 2140 redundant transcripts.
  • the total number of probe sets corresponding to genes present in this list was 3717.
  • IRITs The systemic effect of graft transplantation on IRITs expression also was studied by analyzing IRIT expression in iso-host Dl and D2 kidneys.
  • IRIT-host transcripts One hundred and twenty- nine IRIT-host transcripts were identified that were expressed both in the isografts and in the host kidneys. Expression of these genes probably reflects the systemic effects of surgical procedure. Expression of an additional 17 transcripts was attributed to macrophages. IRITs were annotated using the GO terms. Excluding the parent terms, IRITs were significantly overrepresented in biological processes such as response to stress (including response to wounding and wound healing), cell cycle and cell proliferation, cell communication including cell adhesion, organ development, and morphogenesis. IRITs also were highly represented in extracellular matrix components (including collagens), cytoskeleton and cell junctions.
  • the IRIT expression profile showed a high negative correlation (-0.75) with epithelial transporters.
  • published expression data sets derived from developing kidneys were reanalyzed and compared with the IRITs (Schmidt-Ott et al. (2005) J. Am. Soc. Nephrol. 16:1993-2002; Schwab et al. (2003) Kidney Int.
  • E12.5 uteretic bud, 88 IRITs were identified in E12.5 uteretic bud vs E12.5 metanephron mesenchyme, 65 in combined embryonic kidney tissues stages vs. adult kidney (excluding mesenchyme), and 67 in El 1.5 metanephron mesenchyme vs. adult kidney.
  • Example 5 Gamma Interferon Suppressed Transcripts (GST) Interferon-gamma (IFN- ⁇ ) has a surprising protective effect in organ allografts, in that mouse kidney allografts lacking IFN- ⁇ effects manifest accelerated congestion and necrosis. To understand this protection, histology, inflammatory infiltrate, and gene expression were assessed in IFN- ⁇ receptor-deficient kidney allografts transplanted into wild-type and various knockout hosts. Early congestion and necrosis in the IFN- ⁇ receptor-deficient allografts was unchanged in B cell deficient hosts, but was completely abrogated in hosts deficient either in perforin or in granzymes A and B.
  • IFN- ⁇ acts through the donor IFN- ⁇ receptors to induce signal that determines which effector mechanisms act in the allograft, inhibiting perforin-granzyme- mediated congestion and necrosis and suppressing alternative inflammation.
  • the transcriptomes of allografts deficient in IFN- ⁇ signaling were compared to
  • Genes associated with the response to stress/wounding included highly expressed Chi313, F13al and Fgg.
  • Genes related to peptidase activity included members of the Mmp (e.g., Mmp9, Mmpl2), Adam, and Serpin families.
  • Cell adhesion process genes included genes associated with pattern recognition, e.g., MgIl and C type lectins (Clec family members 1, 4, 7), and Thbsl.
  • Extracellular matrix components included collagens Col3al and Col5a2, and Timpl .
  • GO annotations of GSTs are shown in Table 11.
  • the most highly expressed GSTs in terms of fold increase were those associated with alternative macrophage activation (AMA), i.e., Argl, Chi313, Mmpl2, and other macrophage and/or neutrophil activities (S100a8, S100a9 and Earl 1). Additional AMA markers among the GSTs were Ear2, MgIl, Mmp9, Mrcl, and Thbsl.
  • the top 30 GSTs included IL-6 and chemokines Cxcl2, Cxcl4, Cxcl7, Ccl6, Ccl24. Expression of plasminogen activator inhibitors Serpinb2 and Serpinel also was very high.
  • the GSTs include genes involved in the macrophage response to activation, proteolysis, response to wounding, and cell adhesion.
  • At least 64 GSTs were associated with kidney necrosis (i.e., their expression was significantly decreased when the necrosis of IFN-K receptor-deficient allografts was averted).
  • the most decreased GSTs were Serpinb2, Cxcl7 and Cleclb.
  • Many of the decreased GSTs are known to be involved in response to stress, injury, and tissue repair (e.g., adrenomedullin/Adm, heme oxygenase/Hmoxl, 116, fibulin/Fbln2, tenascin/Tnc and thrombospondinl/Thbsl, Serpinb2, and Serpinel).
  • Example 6 Class I Suppressed Transcripts (CIST)
  • IFN- ⁇ acting on allograft IFN- ⁇ receptors induces a signal that prevents early congestion and necrosis and determines inflammatory phenotype as the alloimmune response develops. It was hypothesized that this signal may be high expression of donor MHC class Ia and Ib proteins, which have the potential to control host infiltrating cells via inhibitory receptors. Thus, it was postulated that class I-deficient allografts should resemble IFN- ⁇ receptor deficient allografts.
  • class I deficient allografts Two types were studied: Tapl transporter-deficient or beta 2 microglobulin-deficient, transplanted into wild-type hosts. Although many IFN- ⁇ - induced transcripts were increased, class I-deficient allografts developed congestion and necrosis between days 5 and 7, similar to IFN- ⁇ receptor-deficient allografts. Expression of TH2 cytokines IL-4 and IL- 13 also was increased, despite abundant IFN- ⁇ expression. Microarray analysis of gene expression identified 78 transcripts elevated in class I- deficient allografts that were previously identified as elevated in IFN- ⁇ -deficient allografts, including many markers of alternative macrophage activation (e.g., arginase 1). Thus, it was proposed that in organ allografts, elevated expression of donor class I induced by IFN- ⁇ delivers an inhibitory signal to host inflammatory cells that prevents early graft necrosis, and also prevents some TH2 type inflammatory features.
  • the transcriptomes of Tap IKO and B2mK0 allografts at day 7 were compared to WT (B6) allografts at day 7 and normal B6 control kidneys. These lists were then corrected for CAT, GRIT, and transcripts showing strain differences, using all probe sets corresponding to genes present in these lists. Seventy-eight unique genes were significantly over-expressed in both types of class I-deficient allografts. These were designated as the "class I suppressed transcripts" (CISTs; Table 13, with humanized versions of the mouse genes listed in Table 14).
  • CISTs class I suppressed transcripts
  • the CIST list was analyzed using the GO browser. After excluding parent categories, GO subcategories containing at least 3 CISTs included: response to external stimulus (including Cxcl4, Cxcl7, 116, Hmoxl, F7 and F13al), angiogenesis (e.g., Thbsl), cellular catabolism (e.g., Argl), endopeptidase activity (including Mmpl2, Serpinel and Serpinb2), and carbohydrate binding (e.g., Mrcl). Many CISTs were associated with the extracellular space, including members of the Mmp and Adam families.
  • response to external stimulus including Cxcl4, Cxcl7, 116, Hmoxl, F7 and F13al
  • angiogenesis e.g., Thbsl
  • cellular catabolism e.g., Argl
  • endopeptidase activity including Mmpl2, Serpinel and Serpinb2
  • carbohydrate binding e.g
  • CISTs included Serpinb2, Mmp 12, Argl, interleukins (IL-6, IL-11), and chemokines (Cxcl4, Cxcl7).
  • Some CISTs had been described as macrophage associated. Indeed, it was found that 32 CISTs were highly expressed in primary macrophages, including alternative macrophage activation (AMA) markers, e.g., arginasel (Argl), mannose receptorl (Mrcl), and Mmpl2. Others were linked to both neutrophils and macrophages (e.g., S100a8 and Earl 1).
  • AMA macrophage activation
  • Argl arginasel
  • Mrcl mannose receptorl
  • Mmpl2 Mmpl2
  • Others were linked to both neutrophils and macrophages (e.g., S100a8 and Earl 1).
  • CISTs represent genes involved in macrophage activation, with activities including proteolysis, angiogenesis, and extracellular matrix remodeling.
  • Example 7 Other gene sets and pathways significantly correlate with the orchestrated response depicted by the gene profiles listed in Tables 1-14 Gene profiles and pathways that significantly positively or negatively correlate with the gene sets listed in Tables 1-14 were identified as follows.
  • Table 19 the SIc score (the geometric mean of the ratios of each SIc probeset to that probeset's average value in the 8 controls) for each of the 143 biopsy for cause samples was calculated. The correlation between these 143 values and the 143 scores (again, sample expression to control average expression ratio) for each probeset on the array was calculated. This set of 54,675 correlations was then ordered. Genes with more than one probeset were reduced to a single probeset - that with the highest absolute value for a correlation. All probesets for genes included in the SIc set, as well as unannotated probesets, were removed. Of the remaining probesets, those with the 25 most positive and 25 most negative correlations were selected.
  • the IRIT score (the geometric mean of the ratios of each IRIT probeset to that probeset's average value in the 8 controls) for each of the 143 biopsy for cause samples was calculated.
  • Table 21 All KEGG pathways represented by more than 5 probesets on the chips were selected. Scores for each KEGG pathway were calculated in the same way as were the SIc scores. The correlation between the SIc scores and each of the 177 KEGG scores (across all 143 biopsies for cause) was calculated. This set of 177 correlations was then ordered. The KEGG pathways with the 25 most positive and 25 most negative correlations were selected. Table 22: All KEGG pathways represented by more than 5 probesets on the chips were selected. Scores for each KEGG pathway were calculated in the same way as were the IRIT scores. The correlation between the IRIT scores and each of the 177 KEGG scores (across all 143 biopsies for cause) was calculated. This set of 177 correlations was then ordered. The KEGG pathways with the 25 most positive and 25 most negative correlations were selected
  • the gene set in Table 19 and the gene pathways in Table 21 correlate with the gene profile shown in Tables 1 and 2 (mouse and human Slcs), while the gene set in Table 20 and the gene pathways in Table 22 correlate with the gene profile in Tables 7 and 8 (mouse and human IRITs).
  • Implant biopsies for transcriptome analysis were obtained by taking 18 gauge core samples from donor kidneys. Donor data were collected retrospectively and recipient data prospectively. Renal allografts were biopsied intra-operatively within one hour of revascularization. One core was sent for routine histology. An additional core sample was immediately placed into RNAlater ® (Qiagen) for subsequent RNA extraction. All biopsies were read using conventional renal histopathologic techniques and scored according to the Banff classification (Racusen et al., supra) by two independent renal histopathologists. Delayed graft function (DGF) was defined as the need for dialysis (RRT) within the first week after transplantation.
  • DGF Delayed graft function
  • Individual donor kidney histologic scores were calculated based on the global kidney score (GKS) system (Remuzzi et al, supra).
  • RNA preparation and amplification Total RNA was isolated using the RNeasy ® Mini Kit (QIAGEN, Valencia, CA), and amplified according to Affymetrix ® protocol (Santa Clara, CA) protocol. If the starting input of cRNA was below 2.5 ⁇ g, an additional round of linear amplification was conducted. RNA yields were measured by UV absorbance and RNA quality assessed by Agilent Bioanalyzer.
  • RNA labeling and hybridization to the Affymetrix ® GeneChip microarrays was carried out according to the protocols included in the Affymetrix ® GeneChip Expression Analysis Technical Manual. Analysis of the transcriptome and clinical data: All sample chips, as well as eight nephrectomy controls (for calculating PBT scores) were pooled into one normalization batch and preprocessed using robust multi-chip averaging (RMA), implemented in Bioconductor version 1.7, R version 2.2. An inter-quartile range (IQR) cutoff of 0.5 Iog2 units was then used to filter out probe sets with low variability across the entire dataset.
  • RMA robust multi-chip averaging
  • Hierarchical clustering and principal components analysis were then used to discover clusters within the dataset without any a priori sample classification.
  • Biological pathways were identified using the KEGG-library (Kanehisa et al. (2006) Nucl. Acids Res. 34: 354-357; or World Wide Web at genome.ad.jp/kegg/).
  • PBTs Pathogenesis based transcript sets
  • the selected PBTs included CATs (reflecting T cell burden), GRITs (reflecting IFN-K effects, IRITS and NIRITs (reflecting injury and repair in isografts and allografts, and RTs as well as Slcs (reflecting epithelial integrity of the kidney organ).
  • PCA principal component analysis
  • Example 11 Transcripts differentially expressed between DD and LD
  • 3718 probe sets were found to be differentially expressed at an fdr of 0.01.
  • 1929 probesets showed a significantly higher expression in DD vs LD samples
  • 1789 probesets a significantly lower expression in DD vs LD samples.
  • Transcripts most significantly increased in DD versus LD included fibrinogens FGG, FGB, and FGA; serine proteinase inhibitors SERPINA3 and SERPINAl; lactotransferrin, LTF; superoxide dismutase, SOD2; and lipopolysaccharide binding protein, LBP. These transcripts were more than 5-fold higher in DD samples.
  • Transcripts reduced in DD versus LD kidneys included many related to metabolism of fatty acids and amino acids (lysine, serine, threonine, tryptophane, arginine, proline and alanine); members of the albumin gene family (albumin, ALB; afamin, AFM; group-specific component, GC); and transporters (e.g. amino-acid transporter SLC7A13, the probe set with the lowest transcript level in DD versus LD).
  • Example 12 Transcripts differentially expressed between 'high risk' and 'low risk' DD kidneys
  • Transcripts demonstrating higher expression in the 'High Risk' versus 'Low Risk' groups included genes associated with the immunoglobulin family, e.g., IGKC, IGKVl -5, IGLJ3, IGHG3, IGHGl; collagens and integrms; chemokines including CCL2, 3, 4, 19, and 20; Toll-like receptor signaling, including CCL3, 4, STATl, Ly96, and CD14; antigen processing and presentation, including HLA-DQAl, HLA-DQBl, HLA-DPAl; and renal injury markers such as HAVCRl (KIM-I). Transcripts demonstrating lower expression in the 'High Risk' versus 'Low Risk' groups predominantly included genes related to glucose, fatty acid, and amino acid metabolism.
  • ROC Receiver Operating Characteristic
  • Figure 7 shows ROC curves for individual PBT scores (RTs, tGRITs, mCATs) or PCl scores in predicting DGF status in the 42 DD kidneys.
  • the PCl scores were based on PBTs and on genes that were IQR filtered.
  • Example 14 Many genes in the LD vs. DD and cluster 2 vs. cluster 3 genes sets are members of previously identified Pathogenesis Based Transcript sets (PBTs)
  • PBTs Pathogenesis Based Transcript sets
  • PBT scores are defined as fold-change relative to nephrectomy controls, averaged over all probesets within each PBT.
  • Figure 5 shows P- values from Bayesian t-tests comparing inter-cluster PBT scores. The p-values were corrected using Benjamini and Hochberg's false discovery rate method. Again, Cluster 3 ("high-risk") was subdivided into samples with and without DGF. Studies were then conducted to determine whether these gene sets predicted early function in ROC analysis.
  • Figure 7 shows ROC curves for individual PBT scores (RTs, tGRITs, mCATs) or PCl scores in predicting DGF status in the 42 DD kidneys. The PCl scores were based on PBTs and on genes that were IQR filtered. Thus, the gene sets have predictive value for early function in human kidney transplants.
  • Example 15 Transcript changes correlate with kidney function in human kidney transplant biopsies and with recovery of function
  • the gene sets were assessed for their correlations with function, with change in function, and with recovery 3 months after the biopsy.
  • the analysis includes 136 biopsies for cause.
  • the values shown are the correlation coefficients of the Iog2 of the geomeans for each gene set shown, with the statistical significance of the correlation indicted as dark green (p ⁇ 0.01) or light green p ⁇ 0.05).
  • Example 16 Assessing tissue rejection
  • Epithelial deterioration is a feature of kidney allograft rejection, including invasion by inflammatory cells (tubulitis) and late tubular atrophy.
  • Epithelial changes in CBA mouse kidneys transplanted into B6 or BALB/c wild-type (WT) or CD 103 deficient (CDl 03 ⁇ ' ⁇ ) recipients were studied. Histology was dominated by early interstitial mononuclear infiltration from day 3 and slower evolution of tubulitis after day 7.
  • Epithelial deterioration and tubulitis were associated with increased CD103 + T cells, but kidney allografts rejecting in CD103 7" hosts manifested tubulitis indistinguishable from WT hosts.
  • tubulitis is a late manifestation of loss of epithelial integrity in rejection and may be a consequence rather than a cause of epithelial deterioration.
  • CD103 (Itgae) knockout mice (Schon et al, J. Immunol, 1999; 162(11):6641- 6649) (CD 103 ⁇ ' ⁇ ) received from Dr. C. M. Parker were bred at the University of Maryland. Other mouse strains were from Jackson Laboratory (Bar Harbor, ME).
  • Non-life-supporting renal transplants were performed as described elsewhere (Halloran et al, J. Immunol, 166:7072-7081 (2001)) using wild-type CBA/J (H-2K k ) mice (CBA) as donors and wild-type C57B1/6J (H-2K b ) (B6), BALB/c (H-2D, I-A d ) (Jabs et al, Am. J. Transplant, 2003; 3(12):1501-1509) or CD103 7" (on a BALB/c background) as recipients. Hosts did not receive immunosuppression. Contralateral host kidney and naive CBA kidney served as controls. Kidneys were harvested on days 3, 4, 5, 7, 14, 21, and 42 post transplant, snap- frozen in liquid nitrogen, and stored at -70°C until further analysis. Ischemic acute tubular necrosis
  • Ischemic injury to the kidney was produced by clamping the left renal pedicle for 60 minutes in three wild-type C57B1/6J mice. Mice were sacrificed at day 7, and kidneys were harvested as described elsewhere (Goes et al., Transplantation, 59:565-572 (1995)), snap-frozen in liquid nitrogen, and stored at -70°C until further analysis.
  • Antibodies were obtained as follows. Rat monoclonal antibody to E-cadherin was obtained from Calbiochem-Novabiochem Corporation (San-Diego CA); mouse monoclonal antibody to Ksp-cadherin was obtained from Zymed Laboratories Inc. (San Francisco, CA); HRP-conjugated goat affinity purified F(ab')2 to rat IgG was obtained from ICN Pharmaceuticals, Inc. (Aurora, OH); HRP-conjugated rabbit anti-rat and HRP- conjugated goat anti-mouse antibody were obtained from Jackson Immunoresearch Laboratories Inc.
  • anti-mouse Fc ⁇ RIII/II antibody was obtained from BD Pharmingen (Mississauga, ON, Canada); anti-CD3 ⁇ and anti-CD 103 were obtained from eBioscience (San Diego, CA); and anti-CD4 and anti-CD8 were obtained from BD Pharmingen.
  • Cryostat sections (4 ⁇ m) were incubated with primary antibodies to E-cadherin or Ksp-cadherin or isotype IgG as control (10 ⁇ g/mL; 90 minutes at room temperature), followed by secondary peroxidase-conjugated antibodies (1 mg/mL; 1 :25 dilution; 90 minutes at room temperature). Slides were developed with diaminobenzidine tetrahydrochloride and hydrogen peroxide, and counterstained with hematoxylin. Isotype controls exhibited no immunostaining.
  • Flow cytometry Kidney was minced, placed in 10 mL of PBS containing 2% BSA and 2 mg/mL collagenase (Sigma- Aldrich), and incubated (37°C for 1 hour) with occasional pressing through a syringe plunger. Cells were strained, washed, and resuspended in PBS containing 0.5% FCS. Prior to flow cytometry, Fc receptors were blocked with anti- mouse Fc ⁇ RIII/II antibody, and IxIO 6 cells were stained using anti-CD3 ⁇ , anti-CD103, anti-CD4, and anti-CD8 antibodies (diluted in 0.5% FCS/PBS).
  • RNA was extracted using CsCl density gradient. Two micrograms of RNA were transcribed using M-MLV reverse transcriptase and random primers. For laser capture microdissection (LCM), frozen sections (8 ⁇ m) were stained with the HistoGene LCM Frozen Section Staining kit (Arcturus, Mountain View, CA).
  • Tubules and interstitial material were captured from day 21 transplants with the LCM instrument (Arcturus, Mountain View, CA), and total cellular RNA was extracted from 150 tubules and interstitial areas using the PicoPure RNA isolation kit (Arcturus).
  • RNA was reverse transcribed and amplified using the TaqMan One-Step RT- PCR kit (Applied Biosystems, Foster City, CA.) in a multiplex reaction for 48 cycles.
  • TaqMan probe/primer combinations were obtained as assay on demand (Applied Biosystems) (Ksp-Cadherin) or designed using Primer Express software version 1.5 (PE Applied Biosystems) (CD 103 : forward: 5'-CAGGAGACGCCGGACAGT-S ', SEQ ID NO:1; reverse: 5'-CAGGGCAAAGTTGCACTCAA-S', SEQ ID NO:2; probe: 5'-AGG- AAGATGGCACTGAGATCGCTATTGTCC-3' SEQ ID NO:3; E-Cadherin: forward: 5'- CTGCCATCCTCGGAATCCTT-3', SEQ ID NO:4; reverse: 5 ' -TGGCTC A AATC AA- AGTCCTGGT-3', SEQ ID NO:5; probe
  • MLR mixed lymphocyte culture
  • CTL cultured effector lymphocytes
  • RNA extraction, dsDNA and cRNA synthesis, hybridization to MOE430A or MOE430 2.0 oligonucleotide arrays were carried out according to the Affymetrix Technical Manual (See, e.g., Affymetrix Technical Manual, 2003 version downloaded from Affymetrix's website) and as described elsewhere (Einecke et al., Am. J. Transplant., 5(4):651-661 (2005)). Equal amounts of RNA from 3 mice (20-25 ⁇ g each) were pooled for each array. For NCBA, allografts, isografts, and contralateral host kidneys, two replicate chips were analyzed at each time point (two independent pools of 3 mice). Data were normalized and analyzed with Microarray Suite Expression Analysis
  • epithelial transporter transcripts as a reflection of epithelial function (glucose transporters, amino acid transporters, and aquaporins) was analyzed. To identify those that are specific for kidney epithelium, the transporters that were present in normal kidney and had 5-fold lower expression or were absent in MLR or CTL were selected. For those transcripts that were represented by more than one probeset on the array, the probeset with annotation "_at" was selected.
  • Blots were incubated with primary antibodies in 5% albumin-TBST overnight (3 ⁇ g/mL, 4°C), washed with TBST, and incubated with secondary antibodies (1 :5000 in 1% milk/TBST; 1 hour at room temperature). After washing, immune complexes were detected with the ECL reagent (Amersham Biosciences) using Fuji Super RX films. Developed films were scanned using GS-800 densitometer and quantified using Quantity One software (Bio-Rad).
  • the late grafts at days 14, 21, and 42 exhibited severe tubular damage with patchy cortical necrosis (30% of the cortex by day 42).
  • the infiltrate in kidney allografts at days 5, 7, and 21 contained 40-60% CD3 + T cells.
  • the infiltrate was 35-50% CD68 + (macrophages), with late appearance of 5% CD19 + B cells at day 21.
  • Table 23 Host kidneys and isografts at days 5, 7, and 21 appeared normal with no inflammation or tubulitis.
  • CATs cytotoxic T lymphocyte-associated transcripts
  • T cells expressing integrin ⁇ E ⁇ 7 are associated with tubulitis lesions, and ⁇ E ⁇ 7 has been implicated in the pathogenesis of tubulitis.
  • CD103 + effector T cells engage and alter tubular epithelium via CD103/E-cadherin interactions to mediate tubulitis, loss of cadherins, and deterioration of epithelial cell function was examined.
  • gene expression levels for selected transporters were analyzed.
  • Transcript levels were determined by analysis of Affymetrix Genechip MOE430A or MOE430 2.0 and are represented as signal strength for normal kidney (NCBA) and fold change compared to NCBA for wild-type allografts at days 3-42 post transplant, isografts, contralateral host kidneys, ATN kidneys, and cultured lymphocytes (MLR and CTL).
  • Slc5a2 (Sl part of proximal tubulus) and Slc5alO decreased by 60 percent and 78 percent at day 5 and continued to decrease during the course of rejection, while Slc5al (S3 part of proximal tubule) decreased only after day 21.
  • the decrease in isografts was less and was stable or improving at days 7 and 21.
  • transcripts for the glucose transporters in the proximal convoluted tubule (Slc2a2 and Slc5a2), where the majority of glucose re-absorption occurs, were decreased early in the course of rejection.
  • Two transporters in the S3 segment of the proximal tubule were either not affected (Slc2al) or decreased late (Slc5al).
  • neutral amino acid transporters Slc7a7, Slc7a8, Slc7a9, Slc7alO, Slc7al2, Slc7al3, and Slcla4
  • Slc3al a cystine, dibasic, and neutral amino acid transport
  • Slclal a high affinity glutamate transport
  • a neurotransmitter transporter transporter Slc6al3
  • transcripts for all transporters except Slcla4 were decreased early in rejecting transplants (mean expression at day 5: 45 percent ⁇ 17 percent of expression in NCBA) and continued to decrease over time (mean expression at day 42: 22 percent ⁇ 8 percent of expression in NCBA). Slcla4 increased early in rejection (2.3 fold) and decreased after day 21. The change in transcript expression was less in isografts (mean expression at day 5: 80 percent ⁇ 44 percent of NCBA) and recovered by day 21 (100 percent ⁇ 51 percent of NCBA).
  • Aquaporins 1, 2, 3, and 4 were present and highly expressed in normal kidney (Table 27). By day 5, mean expression of these aquaporins decreased to 45 percent ⁇ 11 percent of expression in NCBA and continued to decrease throughout the course of rejection to 24 ⁇ 8 percent by day 42. Aquaporins 1, 2, and 3 were very stable in isografts, contralateral host kidneys, and ATN kidneys. Expression of aquaporin 4 was decreased in Iso D7, in ATN kidney, and in contralateral host kidneys, although to a lesser extent than in rejecting kidneys. Aquaporins 5, 7, and 9 were absent in NCBA and throughout the rejection process.
  • Ksp-cadherin mRNA decreased by 50 percent at day 5 post transplant and remained depressed through day 21 (Figure 16A).
  • Western blots revealed decreased protein level at day 7 (25 percent) and 21 (50 percent) post allograft (Figure 16B).
  • Staining for Ksp-cadherin in normal control kidneys was similar to that for E-cadherin ( Figure 17E).
  • Ksp-cadherin staining intensity was lower at day 7 ( Figure 17F) and greatly diminished and redistributed at day 21 ( Figure 17G), similar to changes in E-cadherin.
  • Epithelial deterioration is T-cell mediated but not dependent on cytotoxicity
  • Renal solute carrier transcripts decreased in allografts and isografts in response to transplant injury (mouse)
  • solute carrier family 25 mitochondrial carrier; ornithine transporter, member 15 2355 0.87 0.89 f glucose transporters
  • NCBA normal CBA kidney
  • Iso CBA CBA isograft in CBA host
  • AUo CBA-B6Nude CBA allograft in B6 host
  • nB6 normal B6 kidney
  • nBalb/c normal Balb/c kidney (wildtype); nBalb/c.
  • GKO normal GKO kidney (Balb/c background); CBA + rIFNK: kidney (CBA) from mouse treated with recombinant IFNK; B6 + rIFNK: kidney (B6) from mouse treated with recombinant IFNK; Balb/c+ rIFNK: kidney (Balb/c) from mouse treated with recombinant IFNK; Iso Balb/c: Balb/c isograft in Balb/c host; Iso Balb/cGKO: GKO isograft in GKO host, both on Balb/c background; AUo Balb/c-B6: Balb/c allograft in B6 host; AUo GKO-GKO: Balb/c.GKO allograft in B6.GKO host; AUo GRKO-B6: CBA.GRKO allograft in B6 host; AUo CBA-Nude: CBA allograft in B6 nude hosts; AUo Balb/c-
  • Renal solute carrier transcripts decreased in allografts and isografts in response to transplant injury (humanized)
  • Renal transcripts decreased in allografts and isografts with injury (humanized)
  • Macrophage associated transcripts (MATs) expressed in isografts - IRIT-MATs (mouse)
  • EGF-like module containing, mucin-like, hormone receptor-like
  • BALB GKO isografts were compared to WT BALB isografts.
  • CMV cytomegalo virus
  • Pentose phosphate pathway Glyoxylate and dicarboxylate metabolism
  • Interstitial infiltrate, graft necrosis, edema and peritubular capillary congestion (PTC) were recorded as a percentage positive of the whole cortex area.
  • Tubulitis was scored as the number of tubules with tubulitis in one tissue cross section (for NCBA, Iso D5, Iso D7, Iso D21, WT D3, WT D4, WT D5, WT D7) or in ten high power fields (WT D14, WT D21, WT D42).
  • Arteritis and venulitis lesions were counted and given as the mean number of involved vessels per kidney section. The numbers shown are mean ⁇ standard deviation.
  • MOE 430A array MOE 430A 2.0 array.
  • Iso isografts
  • WT wildtype B6 hosts
  • Left contralateral host kidney
  • ATN ischemic acute tubular necrosis
  • CTL cultured cytotoxic lymphocytes
  • MLR mixed lymphocyte culture
  • Iso isografts
  • WT wildtype B6 hosts
  • Left contralateral host kidney
  • ATN ischemic acute tubular necrosis
  • CTL cultured cytotoxic lymphocytes
  • MOE 430A array Only those aquaporins that were present in NCBA and had low expression in CTL are represented in this table. Numbers represent signal strength for NCBA and fold changes compared to NCBA for all other experimental groups.
  • MOE 430 2.0 array MOE 430 2.0 array.
  • NCBA normal CBA kidney

Abstract

This document relates to methods and materials involved in detecting tissue injury and/or rejection (e.g., injury and/or rejection of transplanted tissue). For example, this document relates to methods and materials involved in the early detection of kidney tissue injury.

Description

TISSUE REJECTION
TECHNICAL FIELD
This document relates to methods and materials involved in detecting tissue injury such as tissue injury that may occur with organ transplant rejection (alloimmune injury) or non-alloimmune injury. For example, this document relates to methods and materials involved in detecting tissue rejection.
BACKGROUND The transplantation of tissue from one human to another has been used for years to save lives and to improve the quality of lives. The first successful kidney transplant was performed in the mid-1950s between identical twin brothers. Since then, donors have grown to include close relatives, distant relatives, friends, and total strangers. In some cases, the recipient may reject the transplanted tissue. Thus, tissue rejection and tissue injury that may be due to alloimmune or non-alloimmune events is a concern for any recipient of transplanted tissue. If a clinician is able to recognize early signs of tissue rejection, anti-rejection drugs and other medication often can be used to reverse tissue rejection and manage injury. Further, understanding molecular mechanisms of injury and rejection will lead to development of improved diagnostics and therapeutics. The success of organ transplantation is limited by the degree of injury resulting from the transplantation process (non-alloimmune injury), and by injury resulting from rejection (the alloimmune response). In kidney transplantation, the renal tubular epithelium is a key target of rejection. Changes in the epithelium have diagnostic significance in T cell mediated renal allograft rejection (TCMR). Entry of mononuclear inflammatory cells into the renal tubular epithelium during TCMR (Racusen et al. (1999) Kidney Int. 55:713-723) is associated with deterioration of renal function (Solez et al. (1993) Kidney Int. 43:1058-1067; and Solez et al. (1993) Kidney Int. 44:411-422). Tubulitis, associated with interstitial infiltration by mononuclear cells, is the principal lesion used to diagnose TCMR using the Banff schema (a pathology diagnostic system; Racusen et al. {supra). Kidneys also can be injured by antibody-mediated rejection
(ABMR), the toxic effects of drugs, and through other mechanisms such as viral disease. SUMMARY
This document is based, in part, on the discovery of nucleic acids that are differentially expressed in tissue that is injured as compared to control tissue that is not injured. As such, this document relates to methods and materials involved in detecting tissue injury, such as injury inherent in an organ that is transplanted or is to be transplanted, or injury that occurs with organ transplantation (e.g., alloimmune injury associated with rejection, or non-alloimune injury that can occur, for example, during surgery). For example, this document relates to methods and materials involved in early detection of tissue injury (e.g., tissue injury due to kidney rejection) and the assessment of a mammal's probability of rejecting tissue such as a transplanted organ. This document also relates to methods and materials involved in assessment of tissue quality and performance (e.g., assessment of donor organs for transplantation, prediction of whether an organ is at increased risk for developing delayed graft function (DGF) following transplantation, and assessment of transplanted organs and their potential to recover from alloimmune or non-alloimmune injury).
By analyzing the expression of nucleic acids as disclosed herein, tissue injury can be detected at a time point prior to the emergence of any visually-observable, histological sign of injury (e.g., in kidney tissue, tubulitis, loss of epithelial mass, marked reduction of E-cadherin and Ksp-cadherin, and redistribution to the apical membrane). In some embodiments, expression levels of "injury-and-repair induced transcripts" (IRITs), "not in isografts injury-and-repair induced transcripts" (NIRITs), "gamma-interferon suppressed transcripts" (GSTs), and "class I suppressed transcripts" (CISTs), including, for example, those listed in Tables 5-14, or expression levels of the solute carriers (Slcs) and renal transcripts (RTs) listed in Tables 1-4, can be assessed to determine whether or not tissue is injured, or to distinguish transplanted tissue that is injured from transplanted tissue that is not injured. In some embodiments, the expression level of gene profiles that significantly correlate with the sets referred to in Tables 1-14 (for example the gene set in Table 19 and/or the gene pathways in Table 21 that correlate with the gene profile shown in Tables 1 and 2, or the gene set in Table 20 and/or the gene pathways in Table 22 that correlate with the gene profile in Tables 7 and 8) can be assessed to determine whether or not tissue is injured, or to distinguish transplanted tissue that is injured from transplanted tissue that is not injured.
This document also relates to nucleic acid arrays that can be used to diagnose tissue injury in a mammal. Such arrays can, for example, allow clinicians to diagnose injury in a donor biopsy, diagnose tissue injury in a transplanted organ, or determine the potential for recovery of organ function in a transplanted organ, based on determination of the expression levels of nucleic acids that are differentially expressed in injured and/or rejected tissue as compared to control tissue that is not injured or rejected. The differential expression of such nucleic acids can be detected in injured tissue prior to the emergence of visually-observable, histological signs of tissue injury or rejection, allowing for early diagnosis of patients having injured transplanted tissue. Such diagnosis can help clinicians determine appropriate treatments for those patients. For example, a clinician who diagnoses a patient as having injured transplanted tissue can treat that patient with medication that suppresses tissue rejection and thus injury (e.g., immunosuppressants). In addition, better therapeutics can be developed that will treat or manage injury events.
In one aspect, this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having an injury and repair profile, wherein the presence of the cells indicates that the tissue is injured. The mammal can be a human. The tissue can be from a biopsy. The tissue can be kidney tissue. The tissue can be tissue to be transplanted into a recipient. The tissue can be tissue that has been transplanted into a recipient. The determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides. In another aspect, this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a not-in-isografts injury and repair profile, wherein the presence of the cells indicates that the tissue is injured. The mammal can be a human. The tissue can be from a biopsy. The tissue can be kidney tissue. The tissue can be tissue to be transplanted into a recipient. The tissue can be tissue that has been transplanted into a recipient. The determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
In another aspect, this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a gamma interferon (IFN-K) suppressed profile, wherein the presence of the cells indicates that the tissue is injured. The mammal can be a human. The tissue can be from a biopsy. The tissue can be kidney tissue. The tissue can be tissue to be transplanted into a recipient. The tissue can be tissue that has been transplanted into a recipient. The determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
In another aspect, this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a class I suppressed profile, wherein the presence of the cells indicates that the tissue is injured. The mammal can be a human. The tissue can be from a biopsy. The tissue can be kidney tissue. The tissue can be tissue to be transplanted into a recipient. The tissue can be tissue that has been transplanted into a recipient. The determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
In yet another aspect, this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a renal transcript (RT) profile, wherein the presence of the cells indicates that the tissue is injured. The mammal can be a human. The tissue can be from a biopsy. The tissue can be kidney tissue. The tissue can be tissue to be transplanted into a recipient. The tissue can be tissue that has been transplanted into a recipient. The determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
In another aspect, this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having a solute carrier (SIc) profile, wherein the presence of the cells indicates that the tissue is injured. The mammal can be a human. The tissue can be from a biopsy. The tissue can be kidney tissue. The tissue can be tissue to be transplanted into a recipient. The tissue can be tissue that has been transplanted into a recipient. The determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
This document also features a method for assessing whether a tissue is at risk for delayed graft function (DGF), wherein the method comprises determining whether or not a tissue contains cells having an injury and repair profile, a non-in-isografts injury and repair profile, an IFN-K suppressed profile, a class I suppressed profile, a RT profile, or a SIc profile, wherein the presence of the cells indicates that the tissue is at risk for DGF. The mammal can be a human. The tissue can be from a biopsy. The tissue can be kidney tissue. The tissue can be tissue to be transplanted into a recipient. The tissue can be tissue that has been transplanted into a recipient. The determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
In another aspect, this document features a method for predicting whether a transplanted tissue will recover from injury, wherein the method comprises determining whether or not a tissue contains cells having an injury and repair profile, a non-in- isografts injury and repair profile, an IFN-K suppressed profile, a class I suppressed profile, a RT profile, or a SIc profile, wherein the presence of the cells indicates that the tissue is not likely to recover from injury. The mammal can be a human. The tissue can be from a biopsy. The tissue can be kidney tissue. The tissue can be tissue to be transplanted into a recipient. The tissue can be tissue that has been transplanted into a recipient. The determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides.
In still another aspect, this document features a method for detecting tissue injury, wherein the method comprises determining whether or not a tissue contains cells having an injury and repair correlated profile or an SIc correlated profile, wherein the presence of the cells indicates that the tissue is injured. The mammal can be a human. The tissue can be from a biopsy. The tissue can be kidney tissue. The tissue can be tissue to be transplanted into a recipient. The tissue can be tissue that has been transplanted into a recipient. The determining step can comprise using PCR or a nucleic acid array, or can comprise using immunohistochemistry or an array for detecting polypeptides. This document also features a method for detecting tissue injury, comprising determining whether or not a tissue contains cells having increased activity of biochemical pathways that correlate with an injury and repair profile, with an SIc profile, with a non-in-isografts injury and repair profile, with a gamma interferon suppressed profile, with a class I suppressed profile, or with an RT profile, wherein the presence of the cells indicates that the tissue is injured.
In another aspect, this document features a nucleic acid array comprising at least 20 nucleic acid molecules, wherein each of the at least 20 nucleic acid molecules has a different nucleic acid sequence, and wherein at least 50 percent of the nucleic acid molecules of the array comprise a sequence from nucleic acid selected from the group consisting of the nucleic acids listed in Tables 1-14, 19, and 20. The array can comprise at least 50 nucleic acid molecules, wherein each of the at least 50 nucleic acid molecules has a different nucleic acid sequence. The array can comprise at least 100 nucleic acid molecules, wherein each of the at least 100 nucleic acid molecules has a different nucleic acid sequence. Each of the nucleic acid molecules that comprise a sequence from nucleic acid selected from the group can comprise no more than three mismatches. At least 75 percent of the nucleic acid molecules of the array can comprise a sequence from nucleic acid selected from the group. At least 95 percent of the nucleic acid molecules of the array can comprise a sequence from nucleic acid selected from the group. The array can comprise glass. The at least 20 nucleic acid molecules can comprise a sequence present in a human.
In another aspect, this document features a computer-readable storage medium having instructions stored thereon for causing a programmable processor to determine whether one or more nucleic acids listed in Tables 5-14, and the third column of Table 20 are present in a tissue sample at elevated levels. The computer-readable storage medium can further comprise instructions stored thereon for causing a programmable processor to determine whether one or more of the nucleic acids listed in Tables 5-14, and the third column of 20 is expressed at a greater level in the tissue sample than in a control tissue sample. In another aspect, this document features a computer-readable storage medium having instructions stored thereon for causing a programmable processor to determine whether one or more nucleic acids listed in Tables 1-4 and the third column of Table 19 are present in a tissue sample at decreased levels. The computer-readable storage medium can further comprise instructions stored thereon for causing a programmable processor to determine whether one or more of the nucleic acids listed in Tables 1-4 and the third column of Table 19 is expressed at a lower level in the tissue sample than in a control tissue sample.
In yet another aspect, this document features an apparatus for determining whether a tissue is injured, the apparatus comprising: one or more collectors for obtaining signals representative of the presence of one or more nucleic acids listed in Tables 1-14, 19, and 20 in a sample from the tissue; and a processor for analyzing the signals and determining whether the tissue is injured. The one or more collectors can be configured to obtain further signals representative of the presence of the one or more nucleic acids in a control sample.
In another aspect, this document features a method for detecting tissue rejection. The method comprises, or consists essentially of, determining whether or not tissue transplanted into a mammal contains cells that express a reduced level of a cadherin polypeptide or a transporter polypeptide, wherein the presence of the cells indicates that the tissue is being rejected. The mammal can be a human. The tissue can be kidney tissue. The tissue can be a kidney. The method can comprise determining whether or not the tissue contains cells that express a reduced level of the cadherin polypeptide. The cadherin polypeptide can be an E-cadherin polypeptide or a Ksp-cadherin polypeptide. The method can comprise determining whether or not the tissue contains cells that express a reduced level of the transporter polypeptide. The transporter polypeptide can be selected from the group consisting of Slc2a2, Slc2a4, Slc2a5 Slc5al, Slc5a2, Slc5alO, Slc7a7, Slc7a8, Slc7a9, Slc7alO, Slc7al2, Slc7al3, Slcla4, Slc3al, Slclal, aquaporin 1, aquaporin 2, aquaporin 3, aquaporin 4, ABC transporter (e.g., a member of the ABC transporter polypeptide family), solute carrier, and ATPase polypeptides. The determining step can comprise measuring the level of mRNA encoding the cadherin polypeptide or the transporter polypeptide. The determining step can comprise measuring the level of the cadherin polypeptide or the transporter polypeptide. The method can comprise determining whether or not the tissue contains cells that express the cadherin polypeptide or the transporter polypeptide at a level less than the average level of expression exhibited in cells from control tissue that has not been transplanted. The determining step can comprise determining whether or not a sample contains the cells, wherein the sample comprises cells, was obtained from tissue that was transplanted into the mammal, and was obtained from the tissue within fifteen days of the tissue being transplanted into the mammal.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
The details of one or more embodiments of the invention are set forth in the description and drawings below. Other features, objects, and advantages of the invention will be apparent from the description and the drawings, and from the claims.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG 1 is a depiction of the algorithm used to develop the unique IRIT list. FIG. 2 is a dendrogram for donor (implant) biopsies of 42 deceased donor (DD) and 45 living donor (LD) kidneys. The DIANA dendrogram is based on all 7376 interquartile range- (IQR-) filtered probesets. Black boxes indicate pairs, and arrows indicate delayed graft function (DGF).
FIG 3 is a graph showing principal component analysis (PCA) of the transcriptome of 87 donor (implant) biopsies, based on the same set of 7376 IQR-fϊltered probesets as clustered in Figure 2. L, samples from 45 living donors; D, samples from 42 deceased donors; boxes, kidneys experiencing post-transplant delayed graft function; green, samples in cluster 1 ; orange, samples in cluster 2; black, samples in cluster 3 as shown in Figure 2.
FIG. 4 is a chart showing pathogenesis based transcript (PBT) scores calculated for the 3 clusters shown in Figure 2. Only those probesets passing the non-specific (IQR) filtering step were used to calculate the scores. Cluster 3 ("high-risk") is subdivided into samples with (n=8) and without (n=13) DGF. LD, living donor implants, Cluster 1; low risk, Cluster 2. PBT scores are defined as fold-change relative to the nephrectomy controls, averaged over all probesets within each PBT.
FIG. 5 is a chart showing p-values from Bayesian t-tests comparing inter-cluster PBT scores, p-values have been corrected using Benjamini and Hochberg's false discovery rate method. The Cluster 3 ("high-risk") group has been subdivided into samples with and without DGF.
FIG. 6 is a graph plotting ROC curves for Principal Component 1 (PCl), showing PCA 1 's value in predicting DGF status in the 42 DD kidneys. PCl was based on all probesets passing the IQR-filter, and on all 87 (LD + DD) samples. Solid line, the smoothed-average ROC curve of all 42 leave-one-out cross validated (LOOCV) estimates; horizontal bars, medians in boxplots; dotted lines, non-smoothed individual ROC curves for each of the LOOCV estimates.
FIG. 7 is a graph plotting ROC curves showing individual PBT scores (RTs, tGRITs, and mCATs) and PCl scores in predicting DGF status in the 42 DD kidneys. The PCl scores were based on genes that were both IQR filtered and PBTs. Horizontal bars, medians in boxplots; dotted lines, non-smoothed individual ROC curves for each of the LOOCV estimates.
FIG. 8. is a table showing the correlation of gene sets with function (GFR) at the time of biopsy and 3 months after biopsy.
FIG. 9. is a table showing the correlation of gene sets with the degree of loss of function/GFR before biopsy (all gene sets; center column) and recovery of function/GFR after biopsy (IRITs, GSTs, CISTs; right column).
FIG. 10 is a table showing that the best correlations between renal function (GFR) and gene sets are with the IRITs, particularly with IRITsD3 and IRITsD5. FIG. 11 is a table showing that the best correlations between degree of loss of function/GFR and gene sets are with the IRITs, especially the IRITsD3 and IRITsD5.
FIG. 12: Histology of rejecting kidneys (CBA into B6 transplants; PAS staining). A) Day 5 transplant with periarterial infiltration (magnification 2Ox). B) Day 5 transplant showing no tubulitis (magnification 100X). C) Day 7 allograft showing interstitial infiltration (magnification 2Ox). D) Day 7 transplant with mild tubulitis (magnification 10Ox). E) Day 21 allograft showing interstitial infiltration and edema (magnification 2Ox). F) Day 21 transplant with marked tubulitis (arrows) and distorted tubules (magnification 10Ox). FIG. 13 : Real time RT-PCR analysis of CD 103 mRNA expression. A) normal kidney (NCBA) and allografts (CBA into B6) at days 5, 7, and 21 post transplant. B) NCBA, contralateral CDlOS 7TiOSt kidneys, CBA kidneys rejecting in CD1037" or in wild-type Balb/c hosts at day 21 post transplant. Values are fold changes relative to control kidney (NCBA), expressed as mean ± SE. Assays were done in duplicate. FIG. 14: Histology of allografts rejecting in wild-type (CB A into Balb/c) or
CD1037" (CBA into CD103) hosts at day 21 post transplant. A) Allograft in wild-type host showing interstitial edema, marked tubulitis (arrows) and distorted tubules (PAS staining, magnification 6Ox). B) Allograft in CDl 03-/- host showing interstitial edema, marked tubulitis (arrows) and distorted tubules, indistinguishable from wild-type (PAS staining, magnification 6Ox). C).Electron microscopy of tubulitis lesions in allografts rejecting in wild-type hosts D) Electron microscopy of tubulitis lesions in allografts rejecting in CD1037" hosts. (Lymphocytes within the tubular epithelial cells; lymphocytes in the interstitium; tubular basement membrane).
FIG. 15: Expression of epithelial transporter transcripts (glucose transporters, amino acid transporters, aquaporins) in isografts and rejecting allografts (CBA into B6) at days 5, 7, and 21 post-transplant, determined by Affymetrix microarrays MOE 430A.
FIG. 16: E-cadherin and Ksp-cadherin in rejecting allografts. A) Real time RT- PCR analysis of mRNA expression of cadherins in rejecting kidney (CBA into B6). Values are fold changes relative to control (CBA) kidney, expressed as means ± SE (n=2, three kidneys in each pool). Assays were done in duplicate. B) Western blot analysis of E-cadherin and Ksp-cadherin expression. Fold changes were calculated from the band intensity ratio of Tx (transplant: CBA into B6) versus C (contralateral kidney: B6). Shown are means ± SE, n=3. Basal levels of cadherins did not differ significantly between normal (CBA mice) and contralateral kidneys (B6 mice). C) E-cadherin and Ksp-cadherin mRNA expression in allografts rejecting in wild-type Balb/c (WT) or CD1O3"7" hosts at day 21 post transplant.
FIG. 17: Immunohistochemical staining of E-cadherin and Ksp-cadherin (magnification 10Ox). Arrows show localization of cadherins. At day 7 post transplant, E-cadherin was localized to the basolateral membrane A) in B6 host kidney and B) in rejecting allografts (CBA into B6). At day 21 post transplant, E-cadherin staining was decreased with some redistribution to the apical membrane C) in allografts rejecting in wild-type hosts (CBA into B6) and D) in allografts rejecting in CDl(B"7" hosts (CBA into CD103"A). E) Ksp-cadherin was localized to the basolateral membrane in normal CBA kidney (control). Ksp-cadherin was decreased in rejecting allografts F) in wild-type hosts (CBA into B6) at day 7 post transplant, G) in wild-type hosts (CBA into B6) at day 21 post transplant and H) in CD 103 hosts (CBA into CD 103) at day 21 post transplant.
DETAILED DESCRIPTION
This document provides methods and materials involved in detecting tissue injury (e.g., injury inherent in a tissue to be transplanted, or tissue injury that may occur with organ transplantation, including alloimmune and non-alloimmune injury) and assessing the potential for recovery of organ function. For example, this document provides methods and materials that can be used to determine whether a tissue is injured or susceptible to injury and delay in function. In some cases, a mammal can be diagnosed as having transplanted tissue that is injured (due to rejection or not) or likely to be injured if it is determined that the tissue contains cells that express altered levels of one or more nucleic acid transcripts, as described herein.
As described herein, the expression levels of particular transcripts, including mouse and human "injury-and-repair induced transcripts" (IRITs), "not in isografts injury-and-repair induced transcripts" (NIRITs), "gamma-interferon suppressed transcripts" (GSTs), and "class I suppressed transcripts" (CISTs) can be used to distinguish tissue (e.g., transplanted tissue) that is injured from tissue that is not injured. This document also is based, in part, on the discovery that the expression levels of mouse "cytotoxic T lymphocyte-associated transcripts" (CATs) and "true gamma-interferon dependent and rejection-induced transcripts" (tGRITs) can be used to distinguish tissue (e.g., transplanted tissue) that is being rejected from tissue that is not being rejected as disclosed, for example, in U.S. Publication Nos. 2006/0269948 and 2006/0269949. For example, the expression levels of nucleic acids listed in Tables 5-14 can be assessed in transplanted tissue to determine whether or not that transplanted tissue is injured. In addition, the description provided herein is based, in part, on the discovery that the expression levels of renal transcripts (RTs) such as those listed in Tables 3 and 4, including the solute carriers (Slcs) listed in Tables 1 and 2, can be used to distinguish tissue that is injured (e.g., transplanted tissue that is injured) from uninjured tissue, hi addition, gene lists and pathways have been identified that are significantly positively or negatively correlated with the gene profiles described in Tables 1-14 (e.g., the gene set in Table 19 and/or the gene pathways in Table 21 that correlate with the gene profile shown in Tables 1 and 2, or the gene set in Table 20 and/or the gene pathways in Table 22 that correlate with the gene profile in Tables 7 and 8). These gene sets and pathways can be used to distinguish tissue that is injured from tissue that is not injured.
For example, a mammal can be diagnosed as having transplanted tissue that is injured if it is determined that the tissue contains cells expressing elevated levels of one or more IRITs, NIRITs, GSTs, and/or CISTs, or that express elevated levels one or more of the nucleic acids listed in Tables 5-14. In some embodiments, a mammal can be diagnosed as having transplanted tissue that is injured if it is determined that the tissue contains cells that express reduced levels of one or more Slcs and RTs listed in Tables 1-4. In some cases, a mammal can be diagnosed as having transplanted tissue that is injured if it is determined that the tissue contains cells that express gene lists and/or pathways that are significantly positively or negatively correlated with the gene profiles described in Tables 1-14 (e.g., the gene set in Table 19 and/or the gene pathways in Table 21 that correlate with the gene profile shown in Tables 1 and 2, or the gene set in Table 20 and/or the gene pathways in Table 22 that correlate with the gene profile in Tables 7 and 8). The term "injury and repair-induced transcripts" or "IRITs" refers to transcripts that are increased in isografts at least once between day 1 and day 21, as compared to normal kidney, excluding allogeneic effects as well as T cell-associated, macrophage associated, and IFN-γ inducible transcripts. Thus, IRITs indicate non-alloimmune effects, such as injury caused by surgery or ischemia reperfusion, for example. The ATN model discussed herein demonstrates ischemia reperfusion injury. In some embodiments, an "IRIT" is identified based on expression that is at least two-fold in kidney isografts as compared to normal kidney. Examples of IRITs include, without limitation, the nucleic acids listed in Tables 7-10. Some IRITs, such as those listed in Table 9, also are primary macrophage associated transcripts (MATs). These transcripts indicate non-alloimmune injury involving innate immune responses.
Some gene sets and pathways have been found to be positively or negatively correlated with IRITs. For example, the genes listed in the first column of Table 20 are negatively correlated with IRITs, while the genes listed in the third column of Table 20 are positively correlated with IRITs. Further, the pathways listed in the left column of Table 22 are negatively correlated with IRITs, while the pathways listed in the right column of Table 22 are positively correlated with IRITs. Thus, increased expression of the positively correlated genes listed in Table 20, increased activity of the positively correlated pathways listed in Table 22, decreased expression of the negatively correlated genes listed in Table 20, or decreased activity of the negatively correlated pathways listed in Table 22, can indicate tissue injury (e.g., non-alloimmune injury).
The term "(not in isografts) injury and repair induced transcripts" or "NIRITs" as used herein refers to transcripts that are elevated in kidney allografts vs. isografts at least once between day 1 and day 42 post transplant in WT hosts, excluding transcriptomes of infiltrating T cells, B cells and macrophages, IFN-K inducible genes, cytotoxic T cell associated transcripts, IFN-γ dependent rejection induced transcripts, and transcripts showing strain differences. A "NIRIT" can be identified based on expression that is increased in kidney allografts as compared to control kidneys, but not increased in kidney isografts as compared to control kidneys. Thus, NIRITs indicate injury that occurs in the parenchyma of the kidney (i.e., the transcriptome of the infiltrating cell compartments have been "removed") and is due to an alloimmune response rather than a non- alloimmune response. Examples of NIRITs include, without limitation, the nucleic acids listed in Tables 5 and 6.
Some nucleic acids that are differentially expressed in tissue that is injured as compared to control tissue that is not injured can be nucleic acids that are suppressed by gamma interferon (IFN-γ). The term "IFN-γ suppressed transcripts" or "GSTs" as used herein refers to transcripts that are expressed in IFN-γ receptor deficient kidney allograft tissue at a level that is greater than the level of expression in WT kidney allograft tissue. In some embodiments, for example, a "GST" is identified based on expression that is increased at least two-fold in IFN-γ receptor deficient kidney allograft tissue as compared to the level of expression in WT kidney allograft tissue. GSTs indicate the underlying alternative inflammatory response to alloimmune and non-alloimmune injury. Examples of GSTs include, without limitation, the nucleic acids listed in Tables 11 and 12.
Some nucleic acids can be suppressed by class-I proteins (e.g., MHC class Ia and/or Ib proteins such as the Tapl transporter and beta 2 microglobulin). The term "class I suppressed transcripts" or "CISTs" as used herein refers to transcripts that are expressed in class I protein (e.g., Tapl transporter and beta 2 microglobulin) deficient kidney allograft tissue at a level that is greater than the expression in WT kidney allograft tissue. In some embodiments, for example, a "CIST" is identified based on expression that is increased at least two-fold in class I deficient kidney allograft tissue as compared to the level of expression in WT kidney allograft tissue. CISTs indicate the underlying alternative inflammatory response that occurs to alloimmune and non-alloimmune injury, and demonstrates the involvement of IFN-K in the process. Examples of CISTs include, without limitation, the nucleic acids listed in Tables 13 and 14.
In some embodiments, a nucleic acid can be included in two or more of the categories described herein. For example, some nucleic acids can be considered to be
GSTs and CISTs. Elevated levels of such GST/CIST nucleic acids can indicate injury in allograft transplants, for example.
The RTs listed in Tables 3 and 4 are renal transcripts that are reduced in allografts and isografts with injury. These transcripts reflect non-alloimmune injury due, for example, to surgical stress, ischemia reperfusion, and other causes, as well as ongoing additional injury effects that occur in alloimmune rejection. The Slcs listed in Tables 1 and 2 are renal solute carrier transcripts that are decreased in allografts and isografts with injury. Like the RTs, the Slcs reflect non-alloimmune injury and alloimmune injury. Some gene sets and pathways have been found to be positively or negatively correlated with Slcs. For example, the genes listed in the first column of Table 19 are negatively correlated with Slcs, while the genes listed in the third column of Table 19 are positively correlated with Slcs. Further, the pathways listed in the left column of Table 21 are negatively correlated with Slcs, while the pathways listed in the right column of Table 21 are positively correlated with Slcs. Thus, reduced expression of the positively correlated genes listed in Table 19, reduced activity of the positively correlated pathways listed in Table 21, increased expression of the negatively correlated genes listed in Table 19, or increased activity of the negatively correlated pathways listed in Table 21 can indicate tissue injury (e.g., non-alloimmune injury or alloimmune injury).
Some nucleic acids can be expressed in T lymphocytes. The term "cytotoxic T lymphocyte-associated transcripts" or "CATs" refers to transcripts that are not usually expressed in kidney but are induced in rejection, and that may reflect T cells recruited to the graft. Examples of CATs include, without limitation, the nucleic acids listed in Table 15. These transcripts are diagnostic for allograft rejection and are referred to in co- pending U.S. Publication No. 2006/0269948.
Some nucleic acids can be regulated by IFN-γ and induced by rejection. The term "true interferon gamma dependent and rejection-induced transcripts" or "tGRITs" refers to rejection-induced transcripts that are IFN-γ-dependent in rejection, and also are unique transcripts that are increased at least 2-fold by rIFN-γ. See, co-pending U.S. Publication No. 2006/0269949. Examples of tGRITs include, without limitation, the nucleic acids listed in Table 16, which can be diagnostic for allograft rejection. The term "transcript" as used herein refers to an mRNA identified by one or more numbered Affymetrix probe sets, while a "unique transcript" is an mRNA identified by only one probe set.
In some embodiments, a tissue can be identified as being injured if it is determined that the tissue contains cells having an injury and repair profile, a not-in- isografts injury and repair profile, an IFN-K suppressed profile, or a class I suppressed profile. As used herein, the term "injury and repair profile" refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 7-10 is present at an elevated level.
The term "not-in-isografts injury and repair profile," as used herein, refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 5 and 6 is present at an elevated level.
As used herein, the term "IFN-K suppressed profile" refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 11 and 12 is present at an elevated level.
The term "class I suppressed profile" refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 13 and 14 is present at an elevated level.
In some embodiments, a tissue can be identified as being injured if it is determined that the tissue contains cells having a RT profile or a SIc profile. As used herein, the term "RT profile" refers to a nucleic acid or polypeptide profile in a sample
(e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 3 and 4 is present at a reduced level, and the term "SIc profile" refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or more) of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 1 and 2 is present at an reduced level.
In some embodiments, a tissue can be identified as being injured if it is determined that the tissue contains cells having a quantitative injury and repair profile, a quantitative not-in-isografts injury and repair profile, a quantitative IFN-K suppressed profile, or a quantitative class I suppressed profile. As used herein, the term "quantitative injury and repair profile" refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 7-10 are present at an elevated level. For example, a quantitative human injury and repair profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 8 are present at an elevated level.
The term "quantitative not-in-isografts injury and repair profile" as used herein refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 5 and 6 are present at an elevated level. For example, a human not-in-isografts injury and repair profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 6 are present at an elevated level. The term "quantitative IFN-K suppressed profile" as used herein refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 11 and 12 are present at an elevated level. For example, a human IFN-K suppressed profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 12 are present at an elevated level.
The term "quantitative class I suppressed profile" as used herein refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 13 and 14 are present at an elevated level. For example, a human class I suppressed profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 14 are present at an elevated level.
In some embodiments, a tissue can be identified as being injured if it is determined that the tissue contains cells having a quantitative RT profile, or a quantitative SIc profile. The term "quantitative RT profile" as used herein refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 3 and 4 are present at a reduced level. For example, a quantitative human RT profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 4 are present at a reduced level.
The term "quantitative SIc profile" as used herein refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 1 and 2 are present at a reduced level. For example, a quantitative human SIc profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in Table 2 are present at a reduced level. In some embodiments, a tissue can be identified as being injured if it is determined that the tissue contains cells having an injury and repair positively correlated profile or an injury and repair negatively correlated profile. As used herein, the term "injury and repair positively correlated profile" refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) of the nucleic acids or polypeptides encoded by the nucleic acids listed in column 3 of Table 20 is present at an elevated level. The term "injury and repair negatively correlated profile" refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) of the nucleic acids or polypeptides encoded by the nucleic acids listed in column 1 of Table 20 is present at an elevated level. The presence of an injury and repair positively correlated profile can indicate that a tissue is injured. The presence of an injury and repair negatively correlated profile also can indicate that a tissue is injured. In some embodiments, a tissue can be identified as being injured if it is determined that the tissue contains cells having a quantitative injury and repair positively correlated profile or a quantitative injury and repair negatively correlated profile. The term "quantitative injury and repair positively correlated profile" as used herein refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 20 are present at an elevated level. For example, a quantitative injury and repair positively correlated profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 20 are present at an elevated level. The term "quantitative injury and repair negatively correlated profile" as used herein refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 20 are present at an elevated level. For example, a quantitative injury and repair negatively correlated profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 20 are present at an elevated level. In some embodiments, a tissue can be identified as being injured if it is determined that the tissue contains cells having an SIc positively correlated profile or an SIc negatively correlated profile. As used herein, the term "SIc positively correlated profile" refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 19 is present at a reduced level. The term "SIc negatively correlated profile" refers to a nucleic acid or polypeptide profile in a sample (e.g., a sample of tissue that is transplanted or is to be transplanted) in which one or more than one (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 19 is present at a reduced level. The presence of an SIc positively correlated profile can indicate that a tissue is injured. The presence of an SIc negatively correlated profile also can indicate that a tissue is injured. In some embodiments, a tissue can be identified as being injured if it is determined that the tissue contains cells having a quantitative SIc positively correlated profile or a quantitative SIc negatively correlated profile. The term "quantitative SIc positively correlated profile" as used herein refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 19 are present at a reduced level. For example, a quantitative SIc positively correlated profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in the third column of Table 19 are present at a reduced level. The term "quantitative SIc negatively correlated profile" as used herein refers to a nucleic acid or polypeptide profile in a sample where one tenth or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 19 are present at a reduced level. For example, a quantitative SIc negatively correlated profile can be a nucleic acid or polypeptide profile in a sample where 10%, 12%, 15%, 18%, 20%, 22%, 23%, 25%, 27%, 29%, 30%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100% of the nucleic acids or polypeptides encoded by the nucleic acids listed in the first column of Table 19 are present at a reduced level.
The methods and materials provided herein can be used to detect tissue injury (e.g., tissue rejection) in any mammal, including, without limitation, a human, monkey, horse, dog, cat, cow, pig, mouse, or rat. In addition, the methods and materials provided herein can be used to detect injury of any type of tissue including, without limitation, kidney, heart, liver, pancreas, and lung tissue. For example, the methods and materials provided herein can be used to determine whether or not a human who received a kidney transplant is experiencing injury of the transplanted kidney.
Any type of sample containing cells can be used to determine whether or not transplanted tissue, tissue that is not transplanted, or tissue that is to be transplanted (e.g., donor biopsy) contains cells that express one or more IRITs, NIRITs, GSTs, and or CISTs, or that express one or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 5-14, at elevated levels. Similarly, any type of sample containing cells can be used to determine whether or not transplanted tissue, tissue that is not transplanted, or tissue that is to be transplanted (e.g., donor biopsy) contains cells that express one or more of the nucleic acids or polypeptides encoded by the nucleic acids listed in Tables 1-4 at decreased levels. Further, any type of sample containing cells can be used to determine whether transplanted tissue, tissue that is not transplanted, or tissue that is to be transplanted (e.g., donor biopsy) contains cells that express one or more nucleic acids that significantly positively or negatively correlate with nucleic acids listed in Tables 1-14. For example, biopsy (e.g., punch biopsy, aspiration biopsy, excision biopsy, needle biopsy, or shave biopsy), tissue section, lymph fluid, blood, and synovial fluid samples can be used, hi some embodiments, a tissue biopsy sample can be obtained directly from a tissue that has been transplanted or is to be transplanted. In some embodiments, a lymph fluid sample can be obtained from one or more lymph vessels that drain from the tissue. A sample can contain any type of cell including, without limitation, cytotoxic T lymphocytes, CD4+ T cells, B cells, peripheral blood mononuclear cells, macrophages, kidney cells, lymph node cells, or endothelial cells. Additional examples of Slcs, RTs, IRITs, NIRITs, GSTs, and CISTs, as well as other transcripts with altered expression levels in injured tissues (e.g., genes in pathways related to glutathione metabolism, fatty acid elongation, and cell communication) can be identified using the procedures described herein. For example, the procedures described in Examples 1 and 2 can be used to identify RTs other than those listed in Tables 1 -4, the procedures described in Examples 1 and 4 can be used to identify IRITs other than those listed in Tables 7-10, the procedures described in Examples 1 and 3 can be used to identify NIRITs other than those listed in Tables 5 and 6, the procedures described in Examples 1 and 5 can be used to identify GSTs other than those listed in Tables 11 and 12, and the procedures described in Examples 1 and 6 can be used to identify CISTs other than those listed in Tables 13 and 14.
The expression of any number of Slcs, RTs, IRITs, NIRITs, GSTs, CISTs, or nucleic acids listed in Tables 1-14, 19, 20, 21, and 22 can be evaluated to determine whether or not transplanted tissue is injured. For example, the expression of one or more than one (e.g., two, three, four, five, six, seven, eight, nine, ten, 15, 20, 25, 30, 40, 50, 75, 100, or more than 100) of the nucleic acids listed in Tables 1-14, 19, 20, 21, and 22 can be used.
The term "elevated level" as used herein with respect to the level of a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 5-14 is any level that is greater than a reference level for that nucleic acid or polypeptide. For example, an elevated level of a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 5-14 can be about 0.3, 0.5, 0.7, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.2, 2.4, 2.6, 2.8, 3, 3.3, 3.6, 4, 4.5, 5, 5.5, 6, 7, 8, 9, 10, 15, 20, or more times greater than the reference level for that nucleic acid or polypeptide, respectively.
The term "reduced level" as used herein with respect to the level of a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 1-4 is any level that is less than a reference level for that nucleic acid or polypeptide. For example, a reduced level of a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 1-4 can be about 0.3, 0.5, 0.7, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.2, 2.4, 2.6, 2.8, 3, 3.3, 3.6, 4, 4.5, 5, 5.5, 6, 7, 8, 9, 10, 15, 20, or more times less than the reference level for that nucleic acid or polypeptide, respectively. The term "reference level" as used herein with respect to a nucleic acid or polypeptide encoded by a nucleic acid listed in Tables 1-14 is the level of that nucleic acid or polypeptide typically expressed by cells in tissues that are free of injury. For example, a reference level of a nucleic acid or polypeptide can be the average expression level of that nucleic acid or polypeptide, respectively, in cells isolated from kidney tissue that has not been injured. In addition, a reference level can be any amount. For example, a reference level can be zero. In this case, any level greater than zero would be an elevated level.
Any number of samples can be used to determine a reference level. For example, cells obtained from one or more healthy mammals (e.g., at least 5, 10, 15, 25, 50, 75, 100, or more healthy mammals) can be used to determine a reference level. It will be appreciated that levels from comparable samples are used when determining whether or not a particular level is an elevated or reduced level. For example, levels from one type of cells are compared to reference levels from the same type of cells. In addition, levels measured by comparable techniques are used when determining whether or not a particular level is an elevated level or a reduced level.
Any suitable method can be used to determine whether or not a particular nucleic acid is expressed at a detectable level or at a level that is greater or less than the average level of expression observed in control cells. For example, expression of a particular nucleic acid can be measured by assessing mRNA expression. mRNA expression can be evaluated using, for example, northern blotting, slot blotting, quantitative reverse transcriptase polymerase chain reaction (RT-PCR), real-time RT-PCR, or chip hybridization techniques. Methods for chip hybridization assays include, without limitation, those described herein. Such methods can be used to determine simultaneously the relative expression levels of multiple mRNAs. Alternatively, expression of a particular nucleic acid can be measured by assessing polypeptide levels. For example, polypeptide levels can be measured using any method such as immuno- based assays (e.g., ELISA), western blotting, or silver staining.
The methods and materials provided herein can be used at any time prior to, during, or following tissue transplantation to determine whether or not the tissue is injured, rejected, or likely to be rejected, hi some embodiments, a sample obtained from a donor at any time prior to transplantation can be assessed for the presence of cells expressing elevated levels of a nucleic acid listed in Tables 5-14, decreased levels of a nucleic acid listed in Tables 1-4, or significant alterations in gene profiles that correlate with genes listed Tables 1-14 (such as the gene profiles and pathways referred to in Tables 19, 20, 21, and 22). For example, a sample can be obtained from a donor 1, 2, 3, 4, 5, 6, 7, or more than 7 days prior to transplant, or can be obtained from a donor tissue within hours (e.g., 1, 2, 3, 4, 6, 8, or 12 hours) prior to transplantation. In some cases, a sample obtained from transplanted tissue at any time following the tissue transplantation can be assessed for the presence of cells expressing elevated levels of a nucleic acid listed in Tables 5-14, or decreased levels of a nucleic acid listed in Tables 1-4. For example, a sample can be obtained from transplanted tissue 1, 2, 3, 4, 5, 6, 7, 8, or more hours after the transplanted tissue was transplanted, hi some cases, a sample can be obtained from transplanted tissue one or more days (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 42, or more days) after the transplanted tissue was transplanted. Typically, a sample can be obtained from transplanted tissue 1 to 7 days (e.g., 1 to 3 days, or 5 to 7 days) after transplantation and assessed for the presence of cells expressing elevated levels of one or more IRITs, NIRITs, GSTs, or CISTs, expressing elevated levels of one or more nucleic acids listed in Tables 5-14, expressing decreased levels of one or more transcripts listed in Tables 1-4, or expressing significant alterations in gene profiles that correlate with genes listed Tables 1-14 (such as those gene profiles and/or pathways referred to in Tables 19, 20, 21, and 22).
In some cases, a mammal can be diagnosed as having transplanted tissue that is being rejected if it is determined that the mammal or tissue contains cells that express a reduced level of a cadherin polypeptide or a transporter polypeptide. Any type of sample containing cells can be used to determine whether or not the mammal or transplanted tissue contains cells that express a reduced level of a cadherin polypeptide or a transporter polypeptide. For example, biopsy (e.g., punch biopsy, aspiration biopsy, excision biopsy, needle biopsy, or shave biopsy), tissue section, lymph fluid, blood, and synovial fluid samples can be used. In some embodiments, a tissue biopsy sample can be obtained directly from the transplanted tissue. In some embodiments, a lymph fluid sample can be obtained from one or more lymph vessels that drain from the transplanted tissue. A sample can contain any type of cell including, without limitation, cytotoxic T lymphocytes, CD4+ T cells, B cells, peripheral blood mononuclear cells, macrophages, kidney cells, lymph node cells, or endothelial cells. Examples of cadherin polypeptides include, without limitation, E-cadherin polypeptides, Ksp-cadherin polypeptides, and any other cadherin polypeptide. Examples of transporter polypeptides include, without limitation, Slc2a2, Slc2a4, Slc2a5 Slc5al, Slc5a2, Slc5alO, Slc7a7, Slc7a8, Slc7a9, Slc7alO, Slc7al2, Slc7al3, Slcla4, Slc3al, Slclal, aquaporins (e.g., aquaporin 1, aquaporin 2, aquaporin 3, and aquaporin 4), members of the family of ABC transporters, solute carriers, and ATPases. The expression of any number of polypeptides disclosed herein or nucleic acids encoding such polypeptides can be evaluated to determine whether or not transplanted tissue will be rejected. For example, the expression of one or more than one (e.g., two, three, four, five, six, seven, eight, nine, ten, 15, 20, 25, 30, 40, 50, 75, 100, or more than 100) of the transporter polypeptides provided herein can be used. In some embodiments, determining that a polypeptide is expressed at a reduced level in a sample can indicate that transplanted tissue will be rejected. In some embodiments, transplanted tissue can be evaluated by determining whether or not the tissue contains cells that express one or more cadherin or transporter polypeptides at a level that is less than the average expression level observed in control cells obtained from tissue that has not been transplanted. Typically, a polypeptide can be classified as being expressed at a level that is less than the average level observed in control cells if the expression levels differ by at least 1-fold (e.g., 1.5-fold, 2-fold, 3-fold, or more than 3-fold). Control cells typically are the same type of cells as those being evaluated. In some cases, the control cells can be isolated from kidney tissue that has not been transplanted into a mammal. Any number of tissues can be used to obtain control cells. For example, control cells can be obtained from one or more tissue samples (e.g., at least 5, 6, 7, 8, 9, 10, or more tissue samples) obtained from one or more healthy mammals (e.g., at least 5, 6, 7, 8, 9, 10, or more healthy mammals).
Any appropriate method can be used to determine whether or not a particular polypeptide is expressed at a reduced level as compared to the average level of expression observed in control cells. For example, expression of a particular polypeptide can be measured by assessing mRNA expression. mRNA expression can be evaluated using, for example, northern blotting, slot blotting, quantitative reverse transcriptase polymerase chain reaction (RT-PCR), real-time RT-PCR, or microarray chip hybridization techniques. Methods for microarray chip hybridization assays include, without limitation, those described herein. Such methods can be used to determine simultaneously the relative expression levels of multiple mRNAs. Alternatively, expression of a particular polypeptide can be measured by assessing polypeptide levels. For example, polypeptide levels can be measured using any method such as immuno- based assays (e.g., ELISA and immunohistochemistry), western blotting, or silver staining.
The methods and materials provided herein can be used at any time following a tissue transplantation to determine whether or not the transplanted tissue will be rejected. For example, a sample obtained from transplanted tissue at any time following the tissue transplantation can be assessed for the presence of cells expressing a reduced level of a polypeptide provided herein, hi some cases, a sample can be obtained from transplanted tissue 1, 2, 3, 4, 5, 6, 7, 8, or more hours after the transplanted tissue was transplanted. In some cases, a sample can be obtained from transplanted tissue one or more days (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, or more days) after the transplanted tissue was transplanted. For example, a sample can be obtained from transplanted tissue 2 to 7 days (e.g., 5 to 7 days) after transplantation and assessed for the presence of cells expressing a reduced level of a polypeptide provided herein. Typically, a biopsy can be obtained any time after transplantation if a patient experiences reduced graft function.
As described herein, a decreased expression of transcripts for many epithelium- specific transporters was found before the onset of tubulitis, indicating that the epithelium is an early target of the rejection process despite the fact that the lymphocytes have no apparent contact with the epithelium, hi addition, the results provided herein demonstrate that the epithelium changes in response to rejection before tubulitis and independent of CD 103, cytotoxic molecules, or antibody acting on the graft. Tubulitis and loss of cadherins in kidney allograft rejection can be associated with CD 103 positive cells in the interstitium and epithelium, while not being dependent on CD 103, and can be part of an ongoing tubulointerstitial process.
Ksp-cadherin mRNA and protein were decreased early, before the onset of tubulitis, coincident with interstitial infiltration. These results demonstrate that the decrease in Ksp-cadherin and E-cadherin can be attributed to the response of the epithelium to the inflammatory processes, responses that can permit the entry of inflammatory cells into the epithelium, and if unchecked can culminate in EMT.
While not being limited to any particular mode of action, one model of tubulitis can be as follows. T cell-mediated rejection in the interstitium can induce expression of effectors (e.g., TGF-βl, actins, vimentin, MMP2, collagens, hyaluronic acid, and many others) that can cause the tubule epithelium to change, permitting the interstitial inflammatory cells to enter the epithelium. The effector T cell/macrophage infiltrate can deliver this contact-independent signal to the epithelium via soluble factors or via matrix- or even microcirculation changes. The mechanism by which the interstitial CTL trigger epithelial changes can be that Tgfbl plays a role. Tgfbl is produced by CTL and is expressed in a CTL line and in recently generated allogeneic cultures, and potentially by macrophages and by many cells in the graft. The early increase in Tgfbl in isografts can exaggerate in allografts, and some Tgfbl -inducible transcripts can be greatly increased in rejecting allografts. In addition, TGF-βl can trigger a decrease in cadherin expression and alterations in epithelial function.
This description also provides nucleic acid arrays. The arrays provided herein can be two-dimensional arrays, and can contain at least 10 different nucleic acid molecules (e.g., at least 20, at least 30, at least 50, at least 100, or at least 200 different nucleic acid molecules). Each nucleic acid molecule can have any length. For example, each nucleic acid molecule can be between 10 and 250 nucleotides (e.g., between 12 and 200, 14 and 175, 15 and 150, 16 and 125, 18 and 100, 20 and 75, or 25 and 50 nucleotides) in length. In addition, each nucleic acid molecule can have any sequence. For example, the nucleic acid molecules of the arrays provided herein can contain sequences that are present within the nucleic acids listed in Tables 1-14, 19, and 20. For the purpose of this document, a sequence is considered present within a nucleic acid listed in, for example, Table 1 when the sequence is present within either the coding or non-coding strand. For example, both sense and anti-sense oligonucleotides designed to human Slc39a5 nucleic acid are considered present within Scl39a5 nucleic acid.
Typically, at least 25% (e.g., at least 30%, at least 40%, at least 50%, at least 60%, at least 75%, at least 80%, at least 90%, at least 95%, or 100%) of the nucleic acid molecules of an array provided herein contain a sequence that is (1) at least 10 nucleotides (e.g., at least 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, or more nucleotides) in length and (2) at least about 95 percent (e.g., at least about 96, 97, 98, 99, or 100) percent identical, over that length, to a sequence present within a nucleic acid listed in any of Tables 1-16. For example, an array can contain 100 nucleic acid molecules located in known positions, where each of the 100 nucleic acid molecules is 100 nucleotides in length while containing a sequence that is (1) 30 nucleotides in length, and (2) 100 percent identical, over that 30 nucleotide length, to a sequence of one of the nucleic acids listed in any of Tables 1-14, 19, and 20. A nucleic acid molecule of an array provided herein can contain a sequence present within a nucleic acid listed in any of Tables 1-14, 19, and 20, where that sequence contains one or more (e.g., one, two, three, four, or more) mismatches. The nucleic acid arrays provided herein can contain nucleic acid molecules attached to any suitable surface (e.g., plastic or glass), hi addition, any method can be use to make a nucleic acid array. For example, spotting techniques and in situ synthesis techniques can be used to make nucleic acid arrays. Further, the methods disclosed in U.S. Patent Nos. 5,744,305 and 5,143,854 can be used to make nucleic acid arrays. This description also provides methods and materials involved in determining the potential for recovery of organ function following injury. For example, Figure 8 shows that the SIc, RT, IRIT, GST and CIST gene sets correlate with function (glomerular filtration rate; GFR) at the time of biopsy and at 3 months after the biopsy. Figure 9 shows that gene sets correlate with the degree of loss of function/GFR before the biopsy (SLCs, RT's, IRITs, ST's, CISTs), as well as with recovery of function/GFR after the biopsy (IRITs, GSTs, CISTs). Figures 10 and 11 show that the best correlation between renal function and gene sets are with the IRITs, especially with IRITsD3 and IRITsD5 (refer to Table 7 (mouse) and Table 8 (human)).
This document also provides methods and materials to assist medical or research professionals in determining whether or not a tissue is injured, is at increased risk for developing DGF following transplantation, or is likely to recover from alloimmune or non-alloimmune injury. Medical professionals can be, for example, doctors, nurses, medical laboratory technologists, and pharmacists. Research professionals can be, for example, principle investigators, research technicians, postdoctoral trainees, and graduate students. A professional can be assisted by (1) determining the level of one or more nucleic acids or polypeptides encoded by nucleic acids listed in Tables 1-14, determining the level of a cadherin polypeptide, or determining the level of a transporter polypeptide in a sample, and (2) communicating information about that level to that professional.
Any method can be used to communicate information to another person (e.g., a professional). For example, information can be given directly or indirectly to a professional. In addition, any type of communication can be used to communicate the information. For example, mail, e-mail, telephone, and face-to-face interactions can be used. The information also can be communicated to a professional by making that information electronically available to the professional. For example, the information can be communicated to a professional by placing the information on a computer database such that the professional can access the information. In addition, the information can be communicated to a hospital, clinic, or research facility serving as an agent for the professional.
Computer-readable medium and an apparatus for predicting rejection This disclosure further provides a computer-readable storage medium configured with instructions for causing a programmable processor to determine whether a tissue that has been or is to be transplanted is injured, and/or to determine the potential for recovery of organ function. The determination of whether a tissue is injured can be carried out as described herein; that is, by determining whether one or more of the nucleic acids listed in Tables 5-14 and the third column of Table 20 is detected in a sample (e.g., a sample of the tissue), or expressed at a level that is greater than the level of expression in a corresponding control tissue, or by determining whether one or more of the nucleic acids listed in Tables 1-4 and the third column of Table 19 is expressed at a level that is less than the level of expression in a corresponding control tissue, hi some cases, it can be determined whether a tissue is being rejected by determining whether or not the tissue contains cells that express a reduced level of a cadherin polypeptide or a transporter polypeptide. The processor also can be designed to perform functions such as removing baseline noise from detection signals.
Instructions carried on a computer-readable storage medium (e.g., for detecting signals) can be implemented in a high level procedural or object oriented programming language to communicate with a computer system. Alternatively, such instructions can be implemented in assembly or machine language. The language further can be compiled or interpreted language.
The nucleic acid detection signals can be obtained using an apparatus (e.g., a chip reader) and a determination of tissue injury can be generated using a separate processor (e.g., a computer). Alternatively, a single apparatus having a programmable processor can both obtain the detection signals and process the signals to generate a determination of whether injury is occurring or is likely to occur, hi addition, the processing step can be performed simultaneously with the step of collecting the detection signals (e.g., "realtime"). Any suitable process can be used to determine whether a tissue that has been or is to be transplanted is injured. In some embodiments, for example, a process can include determining whether a pre-determined number (e.g., one, two, three, four, five, six, seven, eight, nine, ten, 15, 20, 25, 30, 40, 50, 75, 100, or more than 100) of the nucleic acids listed in Tables 5-14 and the third column of Table 20 is expressed in a sample (e.g., a sample of transplanted tissue) at a level that is greater than the average level observed in control cells (e.g., cells obtained from tissue that has not been transplanted or is not to be transplanted, or in a control transplanted tissue). If the number of nucleic acids that are expressed in the sample is equal to or exceeds the pre-determined number, the tissue can be determined to be injured and the potential for recovery of organ/tissue function can be determined to be low, depending on the gene sets that are predominantly altered. If the number of nucleic acids that are expressed in the sample is less than the pre-determined number, the tissue can be determined not to be injured. The steps of this process (e.g., the detection, or non-detection, of each of the nucleic acids) can be carried out in any suitable order.
Also provided herein is an apparatus for determining whether a tissue that has been or is to be transplanted is injured. An apparatus for determining whether tissue injury has occurred can include, for example, one or more collectors for obtaining signals from a sample (e.g., a sample of nucleic acids hybridized to nucleic acid probes on a substrate such as a chip) and a processor for analyzing the signals and determining whether rejection will occur. By way of example, the collectors can include collection optics for collecting signals (e.g., fluorescence) emitted from the surface of the substrate, separation optics for separating the signal from background focusing the signal, and a recorder responsive to the signal, for recording the amount of signal. The collector can obtain signals representative of the presence of one or more nucleic acids listed in Tables 1-14, 19, and 20 (e.g., in samples from transplanted and/or non-transplanted tissue). The apparatus further can generate a visual or graphical display of the signals, such as a digitized representation. The apparatus further can include a display. In some embodiments, the apparatus can be portable.
The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.
EXAMPLES Example 1 - Materials and Methods (Mouse Studies)
These studies utilized a mouse kidney allograft model that develops pathologic lesions that are diagnostic in human graft rejection. Basically, a comparison of mouse kidney pathology to the mouse transcriptome was used to guide understanding of the relationship of lesions to transcriptome changes in human rejection.
Mice: Male CBA/J (CBA) and C57B1/6 (B6) mice were obtained from the Jackson Laboratory (Bar Harbor, ME). IFN-γ deficient mice (BALB/c.GKO) and (B6.129S7-IFNγtmlTs; B6.GKO) were bred in the Health Sciences Laboratory Animal Services at the University of Alberta. Mouse maintenance and experiments were in conformity with approved animal care protocols. CBA (H-2K, I-Ak) into C57B1/6 (B6; H- 2KbDb, I-Ab) mice strain combinations, BALB/c.GKO into B6.GKO were studied across full MHC and non-MHC disparities.
Renal transplantation: Renal transplantation was performed as a non life- supporting transplant model. Recovered mice were killed at day 1, 2, 3, 4, 5, 7, 14, 21 or 42 post-transplant. Kidneys were removed, snap frozen in liquid nitrogen and stored at -70°C. No mice received immunosuppressive therapy. Kidneys with technical complications or infection at the time of harvesting were removed from the study.
Acute Tubular Necrosis (ATN) model of ischemia reperfusion injury: The vascular pedicle of the left CBA kidney was clamped for 1 hour, kept moistened with PBS at 37°C and then released. Animals were kept for 7 days and then sacrificed. The detailed procedure was previously published (Takeuchi et al. (2003) J. Am. Soc. Nephrol. 14:2823-2832). Kidneys representing the ATN model were denoted ATN D7. The histology of the ATN kidneys, in which ischemic injury was induced by cross-clamping 7 days earlier, was reported in detail elsewhere (Goes et al. (1995) Transplant 59:565-572). In brief, these kidneys showed severe acute tubular injury with flattening of tubular epithelium, variation in cell size and shape, cellular swelling, loss of PAS positive brush borders, and individual tubular epithelial cell necrosis with denudation of the epithelium from the basement membrane and shedding of granular cellular debris into the tubular lumen. In addition, tubular regenerative changes with nuclear enlargement, prominent nucleoli, and mitotic figures were observed. Kidneys with ATN also showed interstitial edema and a focal minimal interstitial mononuclear cell infiltrate.
Recombinant IFN-γ. rIFN-γ was a generous gift from Dr. T. Stewart at Genentech (South San Francisco, CA).
Microarrays: High-density oligonucleotide GeneChip 430A and 430 2.0 arrays, GeneChip T7-Oligo(dT) Promoter Primer Kit, Enzo BioArray HighYield RNA
Transcript Labeling Kit, IVT Labeling KIT, GeneChip Sample Cleanup Module, IVT cRNA Cleanup Kit were purchased from Affymetrix (Santa Clara, CA). RNeasy Mini Kit was from Qiagen (Valencia, CA), Superscript II, E. coli DNA ligase, E. coli DNA polymerase I, E. coli RNase H, T4 DNA polymerase, 5X second strand buffer, and dNTPs were from Invitrogen Life Technologies. RNA preparation and hybridization: Total RNA was extracted from individual kidneys using the guanidinium-cesium chloride method and purified RNA using the RNeasy Mini Kit (Qiagen). RNA yields were measured by UV absorbance. The quality was assessed by calculating the absorbance ratio at 260 nm and 280 nm, as well as by using an Agilent Bio Analyzer to evaluate 18 S and 28S RNA integrity.
For each array, RNA from 3 mice was pooled. RNA processing, labeling and hybridization to MOE430 2.0 arrays was carried out according to the protocols included in the Affymetrix GeneChip Expression Analysis Technical Manual (available on the World Wide Web at affymetrix.com). cRNA used for Moe 430 2.0 arrays was labeled and fragmented using an IVT Labeling Kit and IVT cRNA Cleanup Kit.
Sample designation: Normal control kidneys were obtained from CBA mice and designated as NCBA. Allografts rejecting in wild type hosts (B6) at day 3 through day 42 post transplant were designated as WT Dl, D2, D3, D4, D5, D7, D14, D21 and D42, respectively. Corresponding isografts were designated Iso Dl, D2, D3, D4, D5, D7, D14, D21 and D42. Kidneys from mice treated with recombinant IFN-γ were designated rlFN- γ. BALB/c-GKO kidneys (deficient in IFN-γ) rejecting in IFN-γ-deficient B6 hosts at day 5 were designated as GKO D5, and corresponding isografts were designated ISO.GKO D5. All samples (each consisting of RNA pooled from 3 mice) were analyzed by the Moe 430 2.0 arrays in duplicates Sample analysis: RMA-based method: raw microarray data was pre-processed using the RMA method (Bioconductor 1.7; R version 2.2). Microarrays (controls and treatments) were preprocessed separately for each mouse strain combination. After preprocessing, data sets were subjected to variance-based filtering i.e. all probe sets that had an inter-quartile range of less than 0.5 (Iog2 units), across all chips, were removed. Filtered data was then used for transcript selection as follows: transcripts had to have a corrected p-value ≥O.Ol, and had to be increased >2-fold vs. appropriate controls. Corrected p-values were calculated using the "limma" package (fdr adjustment method), which uses an empirical B ayes method for assigning significance.
Example 2 - Renal transcripts (RTs) and Solute Carriers (Slcs) The epithelium in mouse kidney allografts was examined for morphologic changes, and the relationship of such changes to immunologic effector mechanisms was defined. Rejecting allografts showed tubulitis, loss of epithelial mass, marked reduction of E-cadherin and Ksp-cadherin and redistribution to the apical membrane, indicating loss of polarity. Tubulitis and other morphologic changes in the epithelium were dependent on host T cells but independent of host perforin (Prfl), granzymes A and B (GzmA/B), CD 103, and B cells. The changes in epithelial morphology likely reflect the effects of the T cell mediated interstitial inflammatory reaction, analogous to delayed type hypersensitivity (DTH). Studies were conducted to explore the hypothesis that the T cell mediated inflammatory process in kidney allograft rejection induces major changes in renal parenchymal cells before histologic lesions such as tubulitis develop. Morphologic lesions (tubulitis, tubular shrinkage, loss of cadherins, and loss of polarity) may be a consequence and late manifestation of the epithelial response to the T cell mediated inflammatory process, which could be reflected in the transcriptome of renal parenchymal cells before histologic lesions develop. Microarrays were used to explore the early transcriptome changes of renal parenchymal cells in mouse allografts and isografts, their relationship to the evolution of histologic lesions such as tubulitis, and their relationship to immunologic effector mechanisms. To analyze expression of transcripts that reflect changes in the epithelium, two sets of transcripts with high expression in normal kidney and low expression in inflammatory cells were selected. As a first set, epithelial transporters were selected because of their well documented importance for renal function. In particular, studies were focused specifically on the family of Slcs because of their extensive annotation. Members of the SIc family flagged "present" in normal kidney and "absent" (default conditions of GeneChip Operating Software 1.2, Affymetrix®) or with 5-fold lower expression in MLR, CTL, macrophages, fibroblasts, B cells, and CD8+ T cells compared to normal kidney were selected. If transcripts were represented by more than one probeset, the probeset with annotation " at" and with the most robust signal in normal kidney was selected. To extend the analysis to other RTs in an unbiased approach regardless of the gene family, all transcripts represented on the array were subjected to variance-based filtering (Bioconductor 1.7; R version 2.2); i.e., all probe sets with an inter-quartile range < 0.5 (Iog2 units) were removed (Bioinformatics and Computational Biology Solutions Using R and Bioconductor, 2005, Gentleman, Carey, Huber, Irizarry, and Dudoit, eds., Springer, New York). Of the remaining probesets, those with a signal > 50 in all normal kidneys and 5x higher expression in normal kidney compared to MLR, CTL, CD8, B cells, primary macrophages, and fibroblasts (corrected p-value < 0.01) were selected. Corrected p-values were calculated using the "limma" package (FDR adjustment method; Smyth (2004) Stat. Appl. Genet. MoI. Biol. 3(l):Article 3).
The T cell infiltrate in allografts was detectable from day 1 , and extended to the interstitium from days 5 to 7 post transplant, but morphologic epithelial changes did not develop until day 7. Transcripts for most Slcs were reduced in both allografts and isografts in response to transplant injury, but the loss was more severe and progressive in allografts and paralleled the development of tubulitis and other histologic lesions in the epithelium. Mouse Slcs are listed in Table 1 ; humanized versions of the mouse Slcs are listed in Table 2. Weighted sum decomposition of the SIc transcript set identified allospecific changes from day 1 and revealed multiple components of the allospecific epithelial response: sustained and progressive loss of transcripts, and lack of a positive response to injury. To assess whether specific functional subsets were affected by the loss of transcripts more than others, SIc subsets with specific biological functions (transporters of glucose, amino acids, organic ions, metal ions, Na, NaHCl, monocarboxyl acids, and mitochondrial transporters) were selectively analyzed. All subgroups showed a strikingly similar expression pattern in both isografts and allografts, respectively, resembling the pattern with loss of transcripts described earlier for the entire SIc set.
To derive a larger view of the effects of the alloimmune response on the kidney parenchymal cells, a more extensive set of renal transcripts (RTs) that was not restricted to specific gene families was defined (n = 991; Tables 3 (mouse) and 4 (human)). Expression of RTs decreased post transplant, with more severe and progressive loss of transcripts in allografts compared to isografts, thus resembling the changes described for SIc transcripts.
Loss of transcripts was not attributable to simple dilution and affected the majority of renal transcripts, representing a selective structured program that leads to loss of at least some products and presumably function. The early changes in the transcriptome of renal parenchymal cells reflect the same mechanisms as the later development of histologic lesions such as tubulitis: loss of renal transcripts was dependent on the alloimmune response and T cells, but independent of IFN-K, Prfl , GzmA, GzmB, and alloantibody. The loss of epithelial transcripts should offer a system for objectively measuring the changes in renal allograft biopsies that can add to the current Banff system of grading morphologic lesions.
Example 3 - (Not in Isografts) Injury and Repair Induced Transcripts (NIRIT)
The expression of genes during the alloresponse alone were investigated, excluding transcriptomes of infiltrating T cells, B cells and macrophages. Genes inducible by IFN-γ and genes activated in the isografts also were excluded.
First, all transcripts increased in at least one of the allograft conditions, i.e., day 1, 2, 3, 4, 5, 7, 14, 21, or 42 post transplant, were selected. This list then was corrected for IRIT (injury and repair induced transcripts - induced in the isografts), CAT (cytotoxic T cell associated transcripts), GRIT (gamma interferon dependent rejection induced transcripts), MAT (macrophage associated transcripts), BAT (B cell associated transcripts including immunoglobulin transcripts), and transcripts showing strain differences, using all probe sets corresponding to genes present in these lists. The final NIRIT list included 714 nonredundant genes (Table 5 lists the mouse genes; Table 6 lists the humanized versions of the mouse genes).
Example 4 - Injury and Repair Induced Transcripts (IRIT) Organs experience many stresses in the transplant procedure independent of the alloimmune response. To characterize the effects of these stresses on the organ, separately from allogeneic effects, global gene expression in mouse kidney isografts was studied. T cell-associated, macrophage associated, and IFN-γ inducible transcripts were excluded. Despite normal histology, expression of 970 "injury-and-repair inducible transcripts" (IRITs) was increased in isografts. Evaluation of host kidneys, acute tubular necrosis (ATN) model, and developing kidneys indicated that IRITs represent footprints of systemic stress, acute tubular injury and dedifferentiation. IRITs showed enrichment in GeneSpring Gene Ontology (GO) categories related to morphogenesis, extracellular matrix, response to stress and cell cycle. The expression pattern of IRITs showed significant correlations with the KEGG pathways, including TGFβ signaling, apoptosis, and cell cycle.
Using K-means clustering, the time course of IRIT expression was de-convoluted into three profiles, designated IRIT-Dl , IRIT-D3 and IRIT-D5, which were characterized by peak expression in particular days post-transplant (refer to Table 7). The IRIT-Dl profile showed enrichment in systemic response and epithelium development, and IRIT- D3 showed enrichment in stress response, epithelium development, and mesenchyme differentiation, while IRIT-D 5 represented stress response, extracellular matrix, cell cycle, TGFβ signaling, epithelial development, and mesenchyme differentiation. Thus, injury from transplant procedures can induce multiple transcriptional programs that reflect healing and repair, which eventually resolve. It is striking that genes representing different pathways share similar expression profiles, implying an orchestrated response to stress. The algorithm for identifying IRITs is shown in Figure 1. Transcripts that were over-expressed in the isografts at days 1-21 post-transplant were selected. This list was corrected for CAT, GRIT, MAT (670, 567 and 3717, respectively), and transcripts showing strain differences (Famulski et al. (2006) Am. J. Transplant. 6:1342-1354), using all probe sets corresponding to genes present in these lists. This selection yielded 790 unique IRIT (Table 7) that were elevated in the isografts and, most probably, represent kidney cell expression. Humanized versions of the mouse IRITs are listed in Table 8. For identification of primary macrophages associated transcripts (MATs), the microarray data was analyzed by the GCOS method (Famulski et al., supra). Transcripts were required to be flagged as present, increased > 5-fold over the NB6 kidneys in at least one of the culture conditions, and have ae raw signal in NB6 and NCBA kidney below 200. The resulting list contained 2140 redundant transcripts. The total number of probe sets corresponding to genes present in this list was 3717.
Through this analysis, elevated expression of an additional 243 IRIT transcripts was attributed to macrophages present in the grafts (Table 9). Genes induced in the ATN model, which is a mouse model for ischemia reperfusion injury also were studied, and those that overlapped with IRITs were selected. As many as 604 transcripts were found in the IRITs list, and were defined as IRIT-ATN (Table 7). Thus, isografts demonstrated gene expression that is highly comparable to that in the ATN model, despite their normal histology. The top 25 IRITs differentiating allografts from isografts at day 1 , day 2, day 3, day 4, day 5, day 7, and day 21 are listed in Table 10.
The systemic effect of graft transplantation on IRITs expression also was studied by analyzing IRIT expression in iso-host Dl and D2 kidneys. One hundred and twenty- nine IRIT-host transcripts were identified that were expressed both in the isografts and in the host kidneys. Expression of these genes probably reflects the systemic effects of surgical procedure. Expression of an additional 17 transcripts was attributed to macrophages. IRITs were annotated using the GO terms. Excluding the parent terms, IRITs were significantly overrepresented in biological processes such as response to stress (including response to wounding and wound healing), cell cycle and cell proliferation, cell communication including cell adhesion, organ development, and morphogenesis. IRITs also were highly represented in extracellular matrix components (including collagens), cytoskeleton and cell junctions.
Studies then were conducted to investigate which pathways correlate with the IRITs expression profile in isografts at days 1-21. Spearman correlation of IRIT expression profile with the MAPP and KEGG pathways demonstrated high similarity (> 0.75) of 27 pathways, including apoptosis, cell cycle regulation, and TGFβl signaling.
Interestingly, the IRIT expression profile showed a high negative correlation (-0.75) with epithelial transporters. Prompted by enrichment of the GO categories related to morphogenesis and organ development, published expression data sets derived from developing kidneys were reanalyzed and compared with the IRITs (Schmidt-Ott et al. (2005) J. Am. Soc. Nephrol. 16:1993-2002; Schwab et al. (2003) Kidney Int. 64: 1588- 1604.) Genes involved in kidney development were derived using three comparisons: E 12.5 metanephron mesenchyme vs E 12.5 uteretic bud (combined stalk and tip), El 1.5 metanephron mesenchyme vs adult kidney, and combined embryonic kidney tissues stages El 1.5, El 2.5, El 3.5 and El 6.5 vs. adult kidney, excluding metanephron mesenchyme. The IRITs expressed during development were identified using the nonredundant IRITs list and all probe sets corresponding to genes identified in developing kidneys. Eighty four IRITs were identified in E12.5 metanephron mesenchyme vs. E12.5 uteretic bud, 88 IRITs were identified in E12.5 uteretic bud vs E12.5 metanephron mesenchyme, 65 in combined embryonic kidney tissues stages vs. adult kidney (excluding mesenchyme), and 67 in El 1.5 metanephron mesenchyme vs. adult kidney.
Example 5 - Gamma Interferon Suppressed Transcripts (GST) Interferon-gamma (IFN-γ) has a surprising protective effect in organ allografts, in that mouse kidney allografts lacking IFN-γ effects manifest accelerated congestion and necrosis. To understand this protection, histology, inflammatory infiltrate, and gene expression were assessed in IFN-γ receptor-deficient kidney allografts transplanted into wild-type and various knockout hosts. Early congestion and necrosis in the IFN-γ receptor-deficient allografts was unchanged in B cell deficient hosts, but was completely abrogated in hosts deficient either in perforin or in granzymes A and B. Thus, congestion and necrosis was independent of antibody but was completely dependent on host perforin and granzymes A and B. Many features of inflammation were altered, with increased neutrophils and increased transcripts for interleukin-4 (IL-4) and interleukin-13 (IL- 13). Microarray analysis revealed increased expression of many IFN-γ-suppressed transcripts associated with alternative macrophage activation, including arginase 1, matrix metalloproteinase 9, and mannose receptor. The altered inflammation was independent of antibody and largely independent of host perforin or granzymes A and B. Thus, in kidney allografts, IFN-γ acts through the donor IFN-γ receptors to induce signal that determines which effector mechanisms act in the allograft, inhibiting perforin-granzyme- mediated congestion and necrosis and suppressing alternative inflammation. The transcriptomes of allografts deficient in IFN-γ signaling were compared to
WT allografts and normal kidneys. The resulting transcript lists then were corrected for CAT, GRIT, and transcripts showing strain differences, using all probe sets corresponding to genes present in these lists. Two hundred and seventeen non-redundant genes were identified that were over-expressed in IFN-γ-deficient allografts (Table 11 ; humanized versions of the mouse genes are listed in Table 12). The GST list was inspected for the most overrepresented categories. After excluding parent categories, GO subcategories containing at least five GSTs included: response to stress (including response to wounding), cell adhesion, peptidase activity (including metalloendopeptidase activity), and extracellular matrix components. Genes associated with the response to stress/wounding included highly expressed Chi313, F13al and Fgg. Genes related to peptidase activity included members of the Mmp (e.g., Mmp9, Mmpl2), Adam, and Serpin families. Cell adhesion process genes included genes associated with pattern recognition, e.g., MgIl and C type lectins (Clec family members 1, 4, 7), and Thbsl. Extracellular matrix components included collagens Col3al and Col5a2, and Timpl . GO annotations of GSTs are shown in Table 11. The most highly expressed GSTs in terms of fold increase were those associated with alternative macrophage activation (AMA), i.e., Argl, Chi313, Mmpl2, and other macrophage and/or neutrophil activities (S100a8, S100a9 and Earl 1). Additional AMA markers among the GSTs were Ear2, MgIl, Mmp9, Mrcl, and Thbsl. The top 30 GSTs included IL-6 and chemokines Cxcl2, Cxcl4, Cxcl7, Ccl6, Ccl24. Expression of plasminogen activator inhibitors Serpinb2 and Serpinel also was very high. Thus, the GSTs include genes involved in the macrophage response to activation, proteolysis, response to wounding, and cell adhesion. At least 64 GSTs were associated with kidney necrosis (i.e., their expression was significantly decreased when the necrosis of IFN-K receptor-deficient allografts was averted). The most decreased GSTs were Serpinb2, Cxcl7 and Cleclb. Many of the decreased GSTs are known to be involved in response to stress, injury, and tissue repair (e.g., adrenomedullin/Adm, heme oxygenase/Hmoxl, 116, fibulin/Fbln2, tenascin/Tnc and thrombospondinl/Thbsl, Serpinb2, and Serpinel).
Example 6 - Class I Suppressed Transcripts (CIST) In mouse kidney allografts, IFN-γ acting on allograft IFN-γ receptors induces a signal that prevents early congestion and necrosis and determines inflammatory phenotype as the alloimmune response develops. It was hypothesized that this signal may be high expression of donor MHC class Ia and Ib proteins, which have the potential to control host infiltrating cells via inhibitory receptors. Thus, it was postulated that class I-deficient allografts should resemble IFN-γ receptor deficient allografts. Two types of class I deficient allografts were studied: Tapl transporter-deficient or beta 2 microglobulin-deficient, transplanted into wild-type hosts. Although many IFN-γ- induced transcripts were increased, class I-deficient allografts developed congestion and necrosis between days 5 and 7, similar to IFN-γ receptor-deficient allografts. Expression of TH2 cytokines IL-4 and IL- 13 also was increased, despite abundant IFN-γ expression. Microarray analysis of gene expression identified 78 transcripts elevated in class I- deficient allografts that were previously identified as elevated in IFN-γ-deficient allografts, including many markers of alternative macrophage activation (e.g., arginase 1). Thus, it was proposed that in organ allografts, elevated expression of donor class I induced by IFN-γ delivers an inhibitory signal to host inflammatory cells that prevents early graft necrosis, and also prevents some TH2 type inflammatory features.
The transcriptomes of Tap IKO and B2mK0 allografts at day 7 were compared to WT (B6) allografts at day 7 and normal B6 control kidneys. These lists were then corrected for CAT, GRIT, and transcripts showing strain differences, using all probe sets corresponding to genes present in these lists. Seventy-eight unique genes were significantly over-expressed in both types of class I-deficient allografts. These were designated as the "class I suppressed transcripts" (CISTs; Table 13, with humanized versions of the mouse genes listed in Table 14).
The CIST list was analyzed using the GO browser. After excluding parent categories, GO subcategories containing at least 3 CISTs included: response to external stimulus (including Cxcl4, Cxcl7, 116, Hmoxl, F7 and F13al), angiogenesis (e.g., Thbsl), cellular catabolism (e.g., Argl), endopeptidase activity (including Mmpl2, Serpinel and Serpinb2), and carbohydrate binding (e.g., Mrcl). Many CISTs were associated with the extracellular space, including members of the Mmp and Adam families. The 30 most increased CISTs included Serpinb2, Mmp 12, Argl, interleukins (IL-6, IL-11), and chemokines (Cxcl4, Cxcl7). Some CISTs had been described as macrophage associated. Indeed, it was found that 32 CISTs were highly expressed in primary macrophages, including alternative macrophage activation (AMA) markers, e.g., arginasel (Argl), mannose receptorl (Mrcl), and Mmpl2. Others were linked to both neutrophils and macrophages (e.g., S100a8 and Earl 1). Thus, CISTs represent genes involved in macrophage activation, with activities including proteolysis, angiogenesis, and extracellular matrix remodeling.
Overlap between the CISTs and the GSTs (transcripts over-expressed in GRKO allografts) was observed. Of 78 unique CISTs, 56 were increased in GRKO allografts day 7. Thus, expression of many transcripts, including Argl, Mmpl2, Mrcl, and Thsbl can be elevated either when the graft lacks IFN-γ signaling, or has decreased expression of class I in the presence of IFN-γ.
Example 7 - Other gene sets and pathways significantly correlate with the orchestrated response depicted by the gene profiles listed in Tables 1-14 Gene profiles and pathways that significantly positively or negatively correlate with the gene sets listed in Tables 1-14 were identified as follows.
Table 19: the SIc score (the geometric mean of the ratios of each SIc probeset to that probeset's average value in the 8 controls) for each of the 143 biopsy for cause samples was calculated. The correlation between these 143 values and the 143 scores (again, sample expression to control average expression ratio) for each probeset on the array was calculated. This set of 54,675 correlations was then ordered. Genes with more than one probeset were reduced to a single probeset - that with the highest absolute value for a correlation. All probesets for genes included in the SIc set, as well as unannotated probesets, were removed. Of the remaining probesets, those with the 25 most positive and 25 most negative correlations were selected.
Table 20: The IRIT score (the geometric mean of the ratios of each IRIT probeset to that probeset's average value in the 8 controls) for each of the 143 biopsy for cause samples was calculated. The correlation between these 143 values and the 143 scores (again, sample expression to control average expression ratio) for each probeset on the array was calculated. This set of 54,675 correlations was then ordered. Genes with more than one probeset were reduced to a single probeset - that with the highest absolute value for a correlation. All probesets for genes included in the IRIT set, as well as unannotated probesets, were removed. Of the remaining probesets, those with the 25 most positive and 25 most negative correlations were selected
Table 21 : All KEGG pathways represented by more than 5 probesets on the chips were selected. Scores for each KEGG pathway were calculated in the same way as were the SIc scores. The correlation between the SIc scores and each of the 177 KEGG scores (across all 143 biopsies for cause) was calculated. This set of 177 correlations was then ordered. The KEGG pathways with the 25 most positive and 25 most negative correlations were selected. Table 22: All KEGG pathways represented by more than 5 probesets on the chips were selected. Scores for each KEGG pathway were calculated in the same way as were the IRIT scores. The correlation between the IRIT scores and each of the 177 KEGG scores (across all 143 biopsies for cause) was calculated. This set of 177 correlations was then ordered. The KEGG pathways with the 25 most positive and 25 most negative correlations were selected
The gene set in Table 19 and the gene pathways in Table 21 correlate with the gene profile shown in Tables 1 and 2 (mouse and human Slcs), while the gene set in Table 20 and the gene pathways in Table 22 correlate with the gene profile in Tables 7 and 8 (mouse and human IRITs).
Example 8 - Materials and Methods (human studies)
Patients and clinical data: Implant biopsies for transcriptome analysis were obtained by taking 18 gauge core samples from donor kidneys. Donor data were collected retrospectively and recipient data prospectively. Renal allografts were biopsied intra-operatively within one hour of revascularization. One core was sent for routine histology. An additional core sample was immediately placed into RNAlater® (Qiagen) for subsequent RNA extraction. All biopsies were read using conventional renal histopathologic techniques and scored according to the Banff classification (Racusen et al., supra) by two independent renal histopathologists. Delayed graft function (DGF) was defined as the need for dialysis (RRT) within the first week after transplantation. The decision to initiate dialysis was at the discretion of the primary transplant nephrologists and transplant surgeons, with no involvement of study investigators. Known risk factors for poor post-transplant function were defined based on extended donor criteria, and other factors predisposing to acute kidney injury (Schold et al. (2005) Am. J. Transplant. 5(4 Pt l):757-765; Swanson et al. (2002) Am. J. Transplant.2:68-75; Port et al. (2002) Transplant. 74:1281-1286; Nyberg et al. (2003)
Am. J. Transplant. 3:715-721; Ojo et al. (1997) Transplant. 63:968-974; Grossberg et al. (2006) Transplant. 81:155-159; Randhawa (2001) Transplant. 71 :1361-1365; and Remuzzi et al. (1999) J. Am. Soc. Nephrol. 10:2591-2598). These risk factors included: donor age > 60 years; percent sclerosed glomeruli (%SG) > 20%; cold ischemia time (CIT) > 24 hours; revascularization time (RVT) > 45 minutes; intra-operative mean arterial pressure (MAP) < 70 mmHg; surgical complications (vasospasm, mottled kidney, and delayed pinking/turgidity); cerebrovascular accident (CVA) as cause of death; donor creatinine > 130 μmol/L; donor large vessel atherosclerosis; renal histopathologic features of fibrointimal thickening and/or vascular disease (as a surrogate marker of donor hypertension); and other renal pathology present on biopsy. Individual donor kidney histologic scores were calculated based on the global kidney score (GKS) system (Remuzzi et al, supra).
RNA preparation and amplification: Total RNA was isolated using the RNeasy® Mini Kit (QIAGEN, Valencia, CA), and amplified according to Affymetrix® protocol (Santa Clara, CA) protocol. If the starting input of cRNA was below 2.5 μg, an additional round of linear amplification was conducted. RNA yields were measured by UV absorbance and RNA quality assessed by Agilent Bioanalyzer.
Microarray processing: RNA labeling and hybridization to the Affymetrix® GeneChip microarrays (human HuI 33 Plus 2.0) was carried out according to the protocols included in the Affymetrix® GeneChip Expression Analysis Technical Manual. Analysis of the transcriptome and clinical data: All sample chips, as well as eight nephrectomy controls (for calculating PBT scores) were pooled into one normalization batch and preprocessed using robust multi-chip averaging (RMA), implemented in Bioconductor version 1.7, R version 2.2. An inter-quartile range (IQR) cutoff of 0.5 Iog2 units was then used to filter out probe sets with low variability across the entire dataset. Hierarchical clustering and principal components analysis (PCA) were then used to discover clusters within the dataset without any a priori sample classification. Biological pathways were identified using the KEGG-library (Kanehisa et al. (2006) Nucl. Acids Res. 34: 354-357; or World Wide Web at genome.ad.jp/kegg/).
Pathogenesis based transcript sets (PBTs) were tested in relation to differentiation of the various groups of implant samples derived from the unsupervised clustering methods. The selected PBTs included CATs (reflecting T cell burden), GRITs (reflecting IFN-K effects, IRITS and NIRITs (reflecting injury and repair in isografts and allografts, and RTs as well as Slcs (reflecting epithelial integrity of the kidney organ).
Standard class comparison methods were used to compare known classes in search of differentially expressed genes. All "adjusted p-values" reported refer to false discovery rates (fdr), e.g., an adjusted p-value of 0.01 signifies that 1% of the probe sets identified as significant at the 0.01 level will, on average, be false positives.
Among the different patient groups, dichotomous variables were compared using the Chi-square test. Continuous variables were compared using the t-test for those variables which were approximately normally distributed, and the nonparametric Mann- Whitney U test for those that were not normally distributed. Glomerular filtration rate (GFR) was estimated using Cockroft Gault equation: (140 - R age) * R lean body weight * R gender) / (72 * R crea * 0.0113).
Example 9 - Unsupervised transcriptome analysis
Eighty-seven consecutive implant biopsies were included in these studies: 42 from 31 deceased donors (DD), including 11 pairs, and 45 from living donors (LD). Of the 42 DD transplant recipients, 10 had DGF, whereas 1 of 45 LD recipients experienced DGF (p=0.003). The mean duration of follow up was 411 ± 188 days. During follow up, two patients with functioning grafts died, and no further grafts were lost, giving a graft survival rate of '91.1%.
From the 54675 probesets represented on the microarray, 7376 probe sets passed the IQR filter. Unsupervised hierarchical cluster analysis of these 7376 probe sets, using DIANA, revealed two major clusters and one solitary outlier (Figure 2). Interestingly, despite the unsupervised nature of the analysis, kidneys were clustered depending on donor origin: the larger cluster on the left was comprised of two subclusters, Cluster 1 (44 LD kidneys) and Cluster 2 (21 DD kidneys); the cluster on the right, Cluster 3, included 21 DD and 1 LD kidney. One patient in Cluster 1 experienced DGF (2.3%), compared to 2 in Cluster 2 (9.5%) and 8 in Cluster 3 (36.4%). The incidence of DGF was significantly different between Clusters 1 and 3 (p < 0.001) and between Clusters 2 and 3 (p < 0.05). The incidence of DGF was not significantly different between Clusters 1 and 2.
The same set of 7376 IQR filtered probesets was subjected to a further unsupervised principal component analysis (PCA). PCA showed strong grouping of LD versus DD kidneys (Figure 3). There was wider scatter within the DD group, indicating greater heterogeneity among the samples. Clusters 2 and 3 were observed to form a continuum across the space of the first two principal components. The single outlier identified in Figure 2 lies to the most extreme left in Figure 3. This patient had the worst outcome of all 87 patients, requiring RRT for 2 months post-transplantation. Thus, both independent methods of unsupervised analysis revealed a good separation of LD from DD samples, indicating that the gene expression pattern seen in the DD samples is associated with function. Thus, the transcripts detect the difference between LD and DD, and detect significant heterogeneity among DD.
Example 10 - Clinical characteristics and functional outcomes The demographics and clinical characteristics of all LD and DD implants are outlined in Table 17. Major differences between LD and DD groups included: more female donors in LD (p=0.004); greater HLA mismatches in DD (p < 0.001); and longer cold ischemia time in DD (p < 0.001). DD kidneys had a greater percent sclerosed glomeruli compared to LD (p=0.037). The global kidney score was higher in DD versus LD kidneys (p=0.036). Overall, 26 kidneys had a global kidney score > 4 (18 DD, 8 LD). As expected, the incidence of DGF was significantly greater in DD kidneys (p=0.003). Among all DD, the significant differences between patients with DGF versus those with IGF included higher recipient age in DGF (p=0.002), fewer HLA mismatches in DGF (p=0.009), and longer revascularization time in DGF (p=0.039). There were no significant differences in other clinical variables, including donor age and gender. Subsequent acute rejection rates and CMV episodes were not different between DGF and IGF groups.
Between the two clusters of DD kidneys, DGF was significantly greater in Cluster 3 (p=0.03). Serum creatinine was significantly higher in Cluster 3 versus Cluster 2 at day 7 (p=0.008). When patients requiring RRT were excluded, however, day 7 creatinine remained higher in Cluster 3, but was not statistically significant (p=0.103). Thus, the heterogeneity detected by the transcripts corresponds with differences in early function.
The differences between these two clusters of DD kidneys were examined to understand the significance of the heterogeneity in the DD. The single LD kidney in Cluster 3 was omitted, to focus exclusively on DD samples. There were no major differences in donor and recipient characteristics between Clusters 2 versus 3, with the exception of more female donors in Cluster 3 (p=0.011). Clinical factors including cold ischemia time, revascularization time, and intra-operative mean arterial pressure were not different. Furthermore, the percent sclerosed glomeruli and the global kidney score were not different. This confirms that the transcript differences were above and beyond any known clinical differences in these kidneys.
The number of renal risk factors experienced by patients in Clusters 2 and 3 were analyzed to assess whether these may explain the differences in clinical outcome (Table 18). The number of patients experiencing renal risk factors in Clusters 2 and 3 was not different (n = 17 Cluster 2, n = 19 Cluster 3). Among all patients with risk factors, the incidence of DGF was significantly greater in Cluster 3 (p < 0.05). This observation suggests enhanced susceptibility to DGF in Cluster 3, despite remarkable similarity of multiple clinical and histological variables with Cluster 2. Cluster 3 therefore constitutes a 'high risk' and Cluster 2 a 'low risk' group for DGF. By 12 months of follow-up, there were no observable differences in renal function between LD versus DD kidneys or between Clusters 2 and 3. Thus, certain transcripts permit an assessment of probability of good early function versus impaired early function.
Example 11 - Transcripts differentially expressed between DD and LD In a comparison between DD and LD samples, 3718 probe sets were found to be differentially expressed at an fdr of 0.01. Altogether, 1929 probesets showed a significantly higher expression in DD vs LD samples, and 1789 probesets a significantly lower expression in DD vs LD samples. Transcripts most significantly increased in DD versus LD included fibrinogens FGG, FGB, and FGA; serine proteinase inhibitors SERPINA3 and SERPINAl; lactotransferrin, LTF; superoxide dismutase, SOD2; and lipopolysaccharide binding protein, LBP. These transcripts were more than 5-fold higher in DD samples. Others included complement components C6, C3, ClR, ClRL; chemokines CXCL2, CXCLl, CXCL3, CCL3, and IL8. Transcripts reduced in DD versus LD kidneys included many related to metabolism of fatty acids and amino acids (lysine, serine, threonine, tryptophane, arginine, proline and alanine); members of the albumin gene family (albumin, ALB; afamin, AFM; group-specific component, GC); and transporters (e.g. amino-acid transporter SLC7A13, the probe set with the lowest transcript level in DD versus LD).
Example 12 - Transcripts differentially expressed between 'high risk' and 'low risk' DD kidneys
Between Clusters 3 and 2, 1051 probe sets ('High Risk - Low Risk' set), were differentially expressed at an fdr of 0.01 : 404 probesets were increased and 647 decreased in Cluster 3 vs. Cluster 2. Transcripts demonstrating higher expression in the 'High Risk' versus 'Low Risk' groups included genes associated with the immunoglobulin family, e.g., IGKC, IGKVl -5, IGLJ3, IGHG3, IGHGl; collagens and integrms; chemokines including CCL2, 3, 4, 19, and 20; Toll-like receptor signaling, including CCL3, 4, STATl, Ly96, and CD14; antigen processing and presentation, including HLA-DQAl, HLA-DQBl, HLA-DPAl; and renal injury markers such as HAVCRl (KIM-I). Transcripts demonstrating lower expression in the 'High Risk' versus 'Low Risk' groups predominantly included genes related to glucose, fatty acid, and amino acid metabolism.
Example 13 - Genes associated with outcomes
Studies were conducted to determine how many genes were significantly associated with the differences between LD and DD, between DD cases in cluster 2 and cluster 3, and between DD cases with DGF and IGF. Surprisingly, it was found that many (3718) probesets differed between DD and LD, and 1051 between DD in cluster 2 (low risk) versus cluster 3 (high risk) (fdr of 0.01). Many of the genes separating these kidneys had previously been identified in the PBT gene sets described herein and in the other patent applications referred to in this document. Thus, the genes separating DD from LD and high risk DD from low risk DD were the genes previously identified as IRITs, NIRITS, mCATs, GRITs, GSTs, CISTs, RTs, and Slcs.
To determine whether such genes could predict the risk of DGF in a particular kidney, Receiver Operating Characteristic (ROC) analysis performed for Principal Component 1 (PCl) was compared to ROC performed for LD-DD and for cluster 2 vs. cluster 3 genes. PCl was based on all probesets that passed the IQR-filter, and on all 87 (LD + DD) samples. The ROC curve shown in Figure 6 indicates the value of PCl in predicting DGF status in the 42 DD kidneys.
Figure 7 shows ROC curves for individual PBT scores (RTs, tGRITs, mCATs) or PCl scores in predicting DGF status in the 42 DD kidneys. The PCl scores were based on PBTs and on genes that were IQR filtered.
Example 14 - Many genes in the LD vs. DD and cluster 2 vs. cluster 3 genes sets are members of previously identified Pathogenesis Based Transcript sets (PBTs) A large proportion of the transcripts in both the DDvsLD and High Risk vs Low Risk sets (clusters 3 vs. 2) were annotated as members of existing PBT gene sets: CATs, tGRITs, oGRITs, IRITs, NIRITs, GSTs, CISTS, RTs, and Slcs. We therefore looked at gene set scores in the LD, cluster 2, and cluster 3 kidneys (figure 4, 5).. PBT scores are defined as fold-change relative to nephrectomy controls, averaged over all probesets within each PBT. Mean PBT gene set scores for Clusters 1 , 2, and 3 were stratified according to the presence or absence of DGF. Only those genes passing the non-specific (IQR) filtering step were used to calculate the scores. Cluster 3 ("high risk") was subdivided into samples with and without DGF. A continuum of severity of renal injury appeared to extend from LD to 'Low Risk' to 'High Risk' kidneys. Within the 'High Risk' group, those with DGF had significantly increased transcript scores for tGRITs, mCATs, IRITs, and NIRITs, compared to those with IGF (Figure 4), reflecting greater injury, gamma interferon effects, and T cell burden. Figure 5 shows P- values from Bayesian t-tests comparing inter-cluster PBT scores. The p-values were corrected using Benjamini and Hochberg's false discovery rate method. Again, Cluster 3 ("high-risk") was subdivided into samples with and without DGF. Studies were then conducted to determine whether these gene sets predicted early function in ROC analysis. Figure 7 shows ROC curves for individual PBT scores (RTs, tGRITs, mCATs) or PCl scores in predicting DGF status in the 42 DD kidneys. The PCl scores were based on PBTs and on genes that were IQR filtered. Thus, the gene sets have predictive value for early function in human kidney transplants.
Example 15 - Transcript changes correlate with kidney function in human kidney transplant biopsies and with recovery of function
The gene sets were assessed for their correlations with function, with change in function, and with recovery 3 months after the biopsy. The analysis includes 136 biopsies for cause. The values shown are the correlation coefficients of the Iog2 of the geomeans for each gene set shown, with the statistical significance of the correlation indicted as dark green (p<0.01) or light green p<0.05).
The results indicate the gene sets correlate with function (GFR) at the time of biopsy and 3 months after the biopsy (Figure 8). Moreover, certain gene sets correlated with the degree of loss of function/GFR before the biopsy (Figure 9) and recovery of function/GFR after the biopsy (Figure 9). The best correlations were with the IRITs, especially the IRITsD3 and IRITsD5 (Figures 10 and 11).
Example 16 - Assessing tissue rejection Epithelial deterioration is a feature of kidney allograft rejection, including invasion by inflammatory cells (tubulitis) and late tubular atrophy. Epithelial changes in CBA mouse kidneys transplanted into B6 or BALB/c wild-type (WT) or CD 103 deficient (CDl 03 ~'~) recipients were studied. Histology was dominated by early interstitial mononuclear infiltration from day 3 and slower evolution of tubulitis after day 7. Epithelial deterioration and tubulitis were associated with increased CD103+ T cells, but kidney allografts rejecting in CD1037" hosts manifested tubulitis indistinguishable from WT hosts. By microarray analysis, reduced expression of renal epithelial transporter transcripts was observed as early as day 3, indicating that renal epithelium in kidney allograft rejection deteriorates before the onset of tubulitis. Expression decreased progressively through day 42. By day 21, E-cadherin and Ksp-cadherin protein expression was reduced and redistributed. Allografts rejecting in hosts deficient in CD 103, perforin, granzyme A and B, or mature B cells exhibited the same epithelial deterioration as WT hosts. These results demonstrate that the alloimmune response induces early molecular changes in the tubular epithelium that precede morphologic changes, and late changes with tubulitis and loss of cadherins, independent of CD 103, cytotoxic molecules, or antibody acting on the graft. These results also demonstrate that tubulitis is a late manifestation of loss of epithelial integrity in rejection and may be a consequence rather than a cause of epithelial deterioration.
Methods and materials Mice
CD103 (Itgae) knockout mice (Schon et al, J. Immunol, 1999; 162(11):6641- 6649) (CD 103 ~'~) received from Dr. C. M. Parker were bred at the University of Maryland. Other mouse strains were from Jackson Laboratory (Bar Harbor, ME).
To confirm that the CD1O3"7" mice were homozygous, PCR on genomic DNA was performed using primer sequences flanking the inserted neomycin resistance gene as described elsewhere (Schon et al, J. Immunol, 162(11):6641-6649 (1999)).
Transplants
Non-life-supporting renal transplants were performed as described elsewhere (Halloran et al, J. Immunol, 166:7072-7081 (2001)) using wild-type CBA/J (H-2Kk) mice (CBA) as donors and wild-type C57B1/6J (H-2Kb) (B6), BALB/c (H-2D, I-Ad) (Jabs et al, Am. J. Transplant, 2003; 3(12):1501-1509) or CD1037" (on a BALB/c background) as recipients. Hosts did not receive immunosuppression. Contralateral host kidney and naive CBA kidney served as controls. Kidneys were harvested on days 3, 4, 5, 7, 14, 21, and 42 post transplant, snap- frozen in liquid nitrogen, and stored at -70°C until further analysis. Ischemic acute tubular necrosis
Ischemic injury to the kidney was produced by clamping the left renal pedicle for 60 minutes in three wild-type C57B1/6J mice. Mice were sacrificed at day 7, and kidneys were harvested as described elsewhere (Goes et al., Transplantation, 59:565-572 (1995)), snap-frozen in liquid nitrogen, and stored at -70°C until further analysis.
Antibodies
Antibodies were obtained as follows. Rat monoclonal antibody to E-cadherin was obtained from Calbiochem-Novabiochem Corporation (San-Diego CA); mouse monoclonal antibody to Ksp-cadherin was obtained from Zymed Laboratories Inc. (San Francisco, CA); HRP-conjugated goat affinity purified F(ab')2 to rat IgG was obtained from ICN Pharmaceuticals, Inc. (Aurora, OH); HRP-conjugated rabbit anti-rat and HRP- conjugated goat anti-mouse antibody were obtained from Jackson Immunoresearch Laboratories Inc. (West Grove, PA); anti-mouse FcγRIII/II antibody was obtained from BD Pharmingen (Mississauga, ON, Canada); anti-CD3ε and anti-CD 103 were obtained from eBioscience (San Diego, CA); and anti-CD4 and anti-CD8 were obtained from BD Pharmingen.
Histology and electron microscopy
For each sample (normal kidneys, isografts, allografts, contralateral host kidneys, and ATN kidneys), frozen tissue sections (2 μm) were stained with periodic acid-Schiff (PAS) and subjected to histologic analysis as described elsewhere (Jabs et al., Am. J. Transplant., 3(12):1501-1509 (2003)). Electron microscopy was performed on glutaraldehyde-fϊxed tissue.
Immunohistochemistry
Cryostat sections (4 μm) were incubated with primary antibodies to E-cadherin or Ksp-cadherin or isotype IgG as control (10 μg/mL; 90 minutes at room temperature), followed by secondary peroxidase-conjugated antibodies (1 mg/mL; 1 :25 dilution; 90 minutes at room temperature). Slides were developed with diaminobenzidine tetrahydrochloride and hydrogen peroxide, and counterstained with hematoxylin. Isotype controls exhibited no immunostaining.
Flow cytometry Kidney was minced, placed in 10 mL of PBS containing 2% BSA and 2 mg/mL collagenase (Sigma- Aldrich), and incubated (37°C for 1 hour) with occasional pressing through a syringe plunger. Cells were strained, washed, and resuspended in PBS containing 0.5% FCS. Prior to flow cytometry, Fc receptors were blocked with anti- mouse FcγRIII/II antibody, and IxIO6 cells were stained using anti-CD3ε, anti-CD103, anti-CD4, and anti-CD8 antibodies (diluted in 0.5% FCS/PBS).
Real-time RT-PCR
Expression of CD 103, E-cadherin, and Ksp-cadherin was assessed by TaqMan real-time RT-PCR. Total kidney RNA was extracted using CsCl density gradient. Two micrograms of RNA were transcribed using M-MLV reverse transcriptase and random primers. For laser capture microdissection (LCM), frozen sections (8 μm) were stained with the HistoGene LCM Frozen Section Staining kit (Arcturus, Mountain View, CA). Tubules and interstitial material were captured from day 21 transplants with the LCM instrument (Arcturus, Mountain View, CA), and total cellular RNA was extracted from 150 tubules and interstitial areas using the PicoPure RNA isolation kit (Arcturus).
Purified RNA was reverse transcribed and amplified using the TaqMan One-Step RT- PCR kit (Applied Biosystems, Foster City, CA.) in a multiplex reaction for 48 cycles. TaqMan probe/primer combinations were obtained as assay on demand (Applied Biosystems) (Ksp-Cadherin) or designed using Primer Express software version 1.5 (PE Applied Biosystems) (CD 103 : forward: 5'-CAGGAGACGCCGGACAGT-S ', SEQ ID NO:1; reverse: 5'-CAGGGCAAAGTTGCACTCAA-S', SEQ ID NO:2; probe: 5'-AGG- AAGATGGCACTGAGATCGCTATTGTCC-3' SEQ ID NO:3; E-Cadherin: forward: 5'- CTGCCATCCTCGGAATCCTT-3', SEQ ID NO:4; reverse: 5 ' -TGGCTC A AATC AA- AGTCCTGGT-3', SEQ ID NO:5; probe: 5'-AGGGATCCTCGCCCTGCTGATTCTGA- TC-3', SEQ ID NO:6). Gene expression was quantified with the ABI prism 7700
Sequence Detection System (Applied Biosystems) as described elsewhere (Takeuchi et al, J. Am. Soc. Nephrol, 2003; 14(11):2823-2832). Data were normalized to HPRT mRNA, and expressed relative to the expression in control (CBA) kidneys.
Microarrays Microarray analysis was performed on normal kidneys (NCBA), CBA into B6 wildtype allografts at days 3, 4, 5, 7, 14, 21, and 42 posttransplant (WTD3 to WTD42), CBA into Balb/c. wildtype and CBA into Balb/c.CD103 allografts at day 21 (CDl 03 D21), CBA into CBA isografts at days 5, 7, and 21 posttransplant (Iso D7 to Iso D21), contralateral B6 host kidneys at day 5, ATN kidneys at day 7, as well as on a mixed lymphocyte culture (MLR) and cultured effector lymphocytes (CTL) (Einecke et al, Am. J. Transplant., 5(4):651-661 (2005)).
RNA extraction, dsDNA and cRNA synthesis, hybridization to MOE430A or MOE430 2.0 oligonucleotide arrays (GeneChip, Affymetrix), washing and staining were carried out according to the Affymetrix Technical Manual (See, e.g., Affymetrix Technical Manual, 2003 version downloaded from Affymetrix's website) and as described elsewhere (Einecke et al., Am. J. Transplant., 5(4):651-661 (2005)). Equal amounts of RNA from 3 mice (20-25 μg each) were pooled for each array. For NCBA, allografts, isografts, and contralateral host kidneys, two replicate chips were analyzed at each time point (two independent pools of 3 mice). Data were normalized and analyzed with Microarray Suite Expression Analysis
5.0 software (Affymetrix) and GeneSpring™ software (Version 6.1 , Silicon Genetics, CA, USA) as described elsewhere (Einecke et al, Am. J. Transplant., 5(4):651-661 (2005)).
Expression of epithelial transporter transcripts as a reflection of epithelial function (glucose transporters, amino acid transporters, and aquaporins) was analyzed. To identify those that are specific for kidney epithelium, the transporters that were present in normal kidney and had 5-fold lower expression or were absent in MLR or CTL were selected. For those transcripts that were represented by more than one probeset on the array, the probeset with annotation "_at" was selected.
Western blots About 40 mg of kidney was homogenized in buffer (0.1% Nonidet P-40, 0.05% sodium deoxycholate, 0.01% SDS, 150 mM NaCl, 40 raM Tris-HCl pH 7.6, 10 mM 2- mercaptoethanol), treated with 60 μg/ml of PMSF (30 minutes on ice) then centrifuged (18,000 x g; 15 minutes). 150 μg of protein (determined by Bradford reagent, Sigma- Aldrich) were run on 7.5% SDS-PAGE mini-gels (Bio-Rad, Mississauga, ON, Canada) and wet-transferred to Hybond C+ membranes (Amersham Biosciences, Baie d'Urfe, QB, Canada). Quality of transfer and evenness of loading was confirmed with Ponceau S (Sigma-Aldrich). Samples were destained in TBST (140 mM NaCl, 40 mM Tris-HCl pH 7.6, 0.1% Tween 20) and blocked with 5% milk-TBST. To preserve E-cadherin epitopes, all solutions contained 10 mM CaCl2. Blots were incubated with primary antibodies in 5% albumin-TBST overnight (3 μg/mL, 4°C), washed with TBST, and incubated with secondary antibodies (1 :5000 in 1% milk/TBST; 1 hour at room temperature). After washing, immune complexes were detected with the ECL reagent (Amersham Biosciences) using Fuji Super RX films. Developed films were scanned using GS-800 densitometer and quantified using Quantity One software (Bio-Rad).
Results
Development of interstitial infiltrate and tubulins
As described elsewhere (Jabs et al., Am. J. Transplant., 3(12): 1501 -1509 (2003); and Einecke et al., Am. J. Transplant., 5(4):651-661 (2005)), allografts exhibited focal periarterial mononuclear infiltrate at day 3 and 4 and interstitial mononuclear infiltration by day 5 (Figure 12A), which increased at day 7 and persisted through day 42. Tubulitis was absent at day 3, 4, and 5 (Figure 12A) and minimal at day 7, with preserved tubule structure. By day 14, 21, and 42, tubulitis was severe with distortion and shrinkage of tubule cross-sections (Figure 12B), accompanied by endothelial arteritis. The late grafts at days 14, 21, and 42 exhibited severe tubular damage with patchy cortical necrosis (30% of the cortex by day 42). By immunostaining, the infiltrate in kidney allografts at days 5, 7, and 21 contained 40-60% CD3+ T cells. At day 21, T cells were present in the interstitium and tubules, with CD3+CD8+ cells exceeding CD3+CD4+ cells by 8 to 1 (34 ± 4 versus 4 ± 2 cells per 10 HPF, n=9). The infiltrate was 35-50% CD68+ (macrophages), with late appearance of 5% CD19+ B cells at day 21. Detailed histology results were summarized (Table 23). Host kidneys and isografts at days 5, 7, and 21 appeared normal with no inflammation or tubulitis.
A set of cytotoxic T lymphocyte-associated transcripts (CATs) was detectable by day 3 and highly expressed by day 5 in rejecting kidneys, with a median signal to 14 percent of that in cultured effector CTL, compared to 4% in isografts and normal kidneys (Einecke et al., Am. J. Transplant., 5(4):651-661 (2005)). Expression of CATs was established before diagnostic lesions and remained remarkably consistent through day 42 despite massive alterations in the pathology, and probably reflects T cells recruited to the graft.
Expression of CD 103 in rejecting allografts
T cells expressing integrin αEβ7 (CD 103) are associated with tubulitis lesions, and αEβ7 has been implicated in the pathogenesis of tubulitis. The possibility that CD103+ effector T cells engage and alter tubular epithelium via CD103/E-cadherin interactions to mediate tubulitis, loss of cadherins, and deterioration of epithelial cell function was examined.
Flow cytometric analysis of lymphocytes isolated from rejecting kidneys at day 21 post transplant revealed that 32.3 ± 13.7 percent of CD4+ cells and 22.0 ± 9.2 percent of CD8+ cells expressed CD 103 (n=3), confirming that CD103+ T cells are present in the rejecting graft (Hadley et al, Transplant, 1999; 67(11):1418-1425). By RT-PCR analysis, CD 103 mRNA increased 4-fold at day 5 and 14-fold at day 7 post transplant (Figure 13A), and remained 12-fold elevated at day 21. Because the CD 103 antibody was unreliable for localizing CDl 03+ cells, the presence of CD 103 mRNA in the epithelium was confirmed using laser capture microdissection. In kidneys with established tubulitis (day 21), CD 103 mRNA was present in tubules, and was at least as abundant (91 -fold) as in the interstitial infiltrate (42-fold) compared to normal kidney.
Absence of CDl 03 does not prevent tubulitis and epithelial deterioration
Renal allografts transplanted into CD1O3"7" hosts at day 21 post transplant were studied. As expected, CD 103 RNA was absent in contralateral host kidneys and allografts in CD1037" hosts (Figure 13B). The histologic findings in allografts rejecting in CD103 hosts were indistinguishable from those in BALB/c wild-type hosts (Figures 14A and 14B), with edema, distortion of tubules, and florid tubulitis. Electron micrographs of the tubulitis lesions in kidneys rejecting in CD 103 ^" hosts versus controls revealed no differences. In both, the intra-epithelial inflammatory cells were observed tightly applied to the basement membranes (Figures 14C and 14D). Semi-quantitative assessment of histologic lesions in CD1O3"7" hosts revealed no differences (Table 24). Expression of CATs correlated highly with that in kidneys rejecting in wild-type hosts (r = 0.94), confirming a similar T cell burden in the graft.
Expression of transporters in rejecting kidney as indicators of epithelial deterioration
To examine epithelial function and integrity in allografts, gene expression levels for selected transporters (glucose transporters, amino acid transporters, and aquaporins) were analyzed. Transcript levels were determined by analysis of Affymetrix Genechip MOE430A or MOE430 2.0 and are represented as signal strength for normal kidney (NCBA) and fold change compared to NCBA for wild-type allografts at days 3-42 post transplant, isografts, contralateral host kidneys, ATN kidneys, and cultured lymphocytes (MLR and CTL).
1. Glucose transporter transcripts Eight facilitated glucose transporters were represented on the chip (Table 25), six of them present in NCBA (Slc2al, Slc2a2, Slc2a4, Slc2a5, Slc2a8, Slc2a9). Slc2al, Slc2a8, and Slc2a9 were excluded because they were highly expressed in lymphocytes and thus not specific for epithelial cells. Slc2a2, Slc2a4, and Slc2a5 had low expression in CTL. These transcripts decreased in rejecting transplants at day 5 by at least 60 percent and continued to decrease during the course of rejection. Their expression in isografts decreased but to a lesser degree than in allografts (14 %, 24 %, and 33 %, respectively) and was stable or recovered after day 5.
Three glucose transporters in the Na+-Glucose-Cotransporter family (Slc5al, Slc5a2, and Slc5alO) actively transport glucose across the apical brush border of kidney epithelial cells. All were present in NCBA with little or no expression in lymphocytes.
Slc5a2 (Sl part of proximal tubulus) and Slc5alO decreased by 60 percent and 78 percent at day 5 and continued to decrease during the course of rejection, while Slc5al (S3 part of proximal tubule) decreased only after day 21. The decrease in isografts was less and was stable or improving at days 7 and 21.
Thus, transcripts for the glucose transporters in the proximal convoluted tubule (Slc2a2 and Slc5a2), where the majority of glucose re-absorption occurs, were decreased early in the course of rejection. Two transporters in the S3 segment of the proximal tubule were either not affected (Slc2al) or decreased late (Slc5al).
2. Amino acid transporter transcripts Of 29 amino acid transporters represented on the array, ten were present in NCBA with low expression in CTL (Table 26). These include neutral amino acid transporters (Slc7a7, Slc7a8, Slc7a9, Slc7alO, Slc7al2, Slc7al3, and Slcla4), Slc3al (a cystine, dibasic, and neutral amino acid transport), Slclal (a high affinity glutamate transport), and a neurotransmitter transporter (Slc6al3). Expression of transcripts for all transporters except Slcla4 was decreased early in rejecting transplants (mean expression at day 5: 45 percent ± 17 percent of expression in NCBA) and continued to decrease over time (mean expression at day 42: 22 percent ± 8 percent of expression in NCBA). Slcla4 increased early in rejection (2.3 fold) and decreased after day 21. The change in transcript expression was less in isografts (mean expression at day 5: 80 percent ± 44 percent of NCBA) and recovered by day 21 (100 percent ± 51 percent of NCBA).
3. Aquaporin transcripts
Aquaporins 1, 2, 3, and 4 were present and highly expressed in normal kidney (Table 27). By day 5, mean expression of these aquaporins decreased to 45 percent ± 11 percent of expression in NCBA and continued to decrease throughout the course of rejection to 24 ± 8 percent by day 42. Aquaporins 1, 2, and 3 were very stable in isografts, contralateral host kidneys, and ATN kidneys. Expression of aquaporin 4 was decreased in Iso D7, in ATN kidney, and in contralateral host kidneys, although to a lesser extent than in rejecting kidneys. Aquaporins 5, 7, and 9 were absent in NCBA and throughout the rejection process. The results for glucose and amino acid transporters and for aquaporins are summarized in Figure 15, illustrating how many epithelial transport transcripts in rejecting kidneys are depressed at day 5 by a mechanism requiring the allo-response, but before the development of significant tubulitis. In allografts rejecting in CD 103 ~'~ recipients, a decreased expression of glucose transporters, amino acid transporters and aquaporins similar to that in wild-type hosts was observed (Table 28), with a correlation coefficient r = 0.84.
Cadherins in rejecting kidneys E-cadherin mRNA levels fell only transiently in rejecting kidney at day 5 (Figure
16A). Western blot analysis confirmed this finding, revealing that E-cadherin protein decreased in rejecting kidney by 40 percent at day 21 compared to the contralateral control kidney (Figure 16B), suggesting that post-transcriptional mechanisms contribute to the reduced E-cadherin staining. By immunostaining, E-cadherin was expressed on the basolateral membrane of tubular epithelial cells in control kidney (CBA) and in the contralateral host kidney at day 7 (Figure 17A) and day 21. All tubules were positive for E-cadherin in the basolateral membrane, although the intensity was highly variable among tubules. In rejecting allografts, staining intensity was unchanged at day 7 post transplant (Figure 17B), but by day 21 E-cadherin staining was both severely decreased and redistributed, with loss of polarity manifested by staining of the luminal membrane and loss of basolateral staining in some tubules (Figure 17C).
Ksp-cadherin mRNA decreased by 50 percent at day 5 post transplant and remained depressed through day 21 (Figure 16A). Western blots revealed decreased protein level at day 7 (25 percent) and 21 (50 percent) post allograft (Figure 16B). Staining for Ksp-cadherin in normal control kidneys was similar to that for E-cadherin (Figure 17E). In rejecting kidney, Ksp-cadherin staining intensity was lower at day 7 (Figure 17F) and greatly diminished and redistributed at day 21 (Figure 17G), similar to changes in E-cadherin.
Comparison of day 21 CBA allografts rejecting either in BALB/c or CD1037" hosts revealed that the decrease in Ksp-Cadherin mRNA and the persistence of E- Cadherin mRNA was similar in both groups (Figure 16C). E-Cadherin and Ksp- Cadherin staining was decreased in the allografts rejecting in CDl 03 ~'~ hosts at day 21, similar to the findings in wild-type hosts (Figure 17D and 17H, respectively).
Epithelial deterioration is T-cell mediated but not dependent on cytotoxicity
A decrease in expression of epithelial transporters and cadherins was observed in kidneys rejecting in hosts lacking perforin, granzyme A and B, or mature B-cells, similar to those in wild-type hosts (Table 29).
Table 1
Renal solute carrier transcripts decreased in allografts and isografts in response to transplant injury (mouse)
Affymetrix ID Gene Gene name NCBA lso CBA lso CBA Symbol D1 D2
Solute carriers a) Metal Ion transport
1429523 a at Slc39a5 solute carrier family 39 (metal ion transporter), member 5 314 0.83 1.25
1416832 at Slc39a8 solute carrier family 39 (metal ion transporter), member 8 872 0.64 0.94
1427339 at Slc30a2 solute carrier family 30 (zinc transporter), member 2 463 0.70 0.71 b) Na dependent transport
1450245 at SId 0a2 solute carrier family 10, member 2 51 0.67 0.80
1431379 a at SId 3a1 solute carrier family 13 (sodium/sulphate symporters), member 1 6785 1.55 1.29
1418857 at SId 3a2 solute carrier family 13 (sodium-dependent dicarboxylate transporter), member 2 624 0.89 1.01
1416560 at SId 3a3 solute carrier family 13 (sodium-dependent dicarboxylate transporter), member 3 2139 0.75 1.03
1417280 at SId 7a1 solute carrier family 17 (sodium phosphate), member 1 6446 0.76 0.84
1418923 at SId 7a3 expressed sequence AW261723 9580 0.62 0.75
1423279 at Slc34a1 solute carrier family 34 (sodium phosphate), member 1 10203 0.68 0.89
1439519 at Slc34a3 solute carrier family 34 (sodium phosphate), member 3 1243 0.46 0.58
1434867 at Slc4a11 solute carrier family 4, sodium bicarbonate transporter-like, member 11 283 1.08 1.35
1439137 at Slc4a9 solute carrier family 4, member 9 250 1.05 0.99
1419057 at Slc5a1 solute carrier family 5 (sodium/glucose cotransporter), member 1 578 0.84 1.06
1440834 at Slc5a10 solute carrier family 5, (sodium/glucose cotransporter) member 10 1530 0.96 0.93
1428752 at Slc5a11 solute carrier family 5 (sodium/glucose cotransporter), member 10 509 0.48 0.79
1437755 at Slc5a12 solute carrier family 5 (sodium/glucose cotransporter), member 12 3255 0.64 0.72
1419166 at Slc5a2 solute carrier family 5 (sodium/glucose cotransporter), member 2 2451 0.57 0.64
1426634 at Slc5a9 solute carrier family 5 (sodium/glucose cotransporter), member 9 117 0.75 0.93 c) Na H or Cl transport
1437259 at Slc9a2 solute carrier family 9 (sodium/hydrogen exchanger), member 2 269 0.72 0.89
1441236 at Slc9a3 solute carrier family 9 (sodium/hydrogen exchanger), member 3 845 0.56 0.73
1439368 a at Slc9a3r2 solute carrier family 9 (sodium/hydrogen exchanger), isoform 3 regulator 2 259 1.66 1.54
1450348 at SId 9a3 solute carrier family 19 (sodium/hydrogen exchanger), member 3 104 0.69 0.89
1421390 at SId 2a1 solute carrier family 12, member 1 23 0.78 0.79
1422856 at SId 2a3 solute carrier family 12, member 3 2557 0.76 0.85 d) monocarbox acid transporters
1418446 at SId 6a2 solute carrier family 16 (monocarboxylic acid transporters), member 2 1243 0.72 0.89
1426082 a at SId 6a4 solute carrier family 16 (monocarboxylic acid transporters), member 4 852 0.48 0.57
1429727 at SId 6a9 solute carrier family 16 (monocarboxylic acid transporters), member 9 2019 0.79 0.83 e) mitochondrial transporters
1420967 at Slc25a15 | solute carrier family 25 (mitochondrial carrier; ornithine transporter), member 15 2355 0.87 0.89 f) glucose transporters
1449067 at Slc2a2 solute carrier family 2 (facilitated glucose transporter), member 2 2999 0.81 0.71
1415958 at Slc2a4 solute carrier family 2 (facilitated glucose transporter), member 4 255 0.80 0.98
1416639 at Slc2a5 solute carrier family 2 (facilitated glucose transporter), member 5 1366 0.47 0.64 g) amino acid transporters
1449301 at Slc7a13 solute carrier family 7, (cationic amino acid transporter, y+ system) member 13 9097 0.37 0.67
1426069 s at Slc7a4 solute carrier family 7 (cationic amino acid transporter, y+ system), member 4 82 0.84 1.02
1417392 a at Slc7a7 solute carrier family 7 (cationic amino acid transporter, y+ system), member 7 3004 1.23 1.06
1448783 at Slc7a9 solute carrier family 7 (cationic amino acid transporter, y+ system), member 9 3313 0.97 0.86
1448299 at SId a1 solute carrier family 1 , member 1 2537 0.66 0.79
1448741 at Slc3a1 solute carrier family 3, member 1 4129 0.81 0.83
1418706 at Slc38a3 solute carrier family 38, member 3 169 1.30 0.83 h) organic ion transport
1418118 at Slc22a1 solute carrier family 22 (organic cation transporter), member 1 4107 0.81 0.88
1422897 at Slc22al2 solute carrier family 22 (organic cation transporter)-like 2 1640 0.71 0.51
1419129 at Slc22a13 solute carrier family 22 (organic cation transporter), member 13 797 0.53 0.74
1448209 a at Slc22a17 solute carrier family 22 (organic cation transporter), member 17 406 0.76 0.90
1417809 at Slc22a1l solute carrier family 22 (organic cation transporter), member 1 -like 4286 0.61 0.71
1425038 at Slc22a19 solute carrier family 22 (organic anion transporter), member 19 1904 0.56 0.63
1419117 at Slc22a2 solute carrier family 22 (organic cation transporter), member 2 3169 0.67 0.66
1417639 at Slc22a4 solute carrier family 22 (organic cation transporter), member 4 341 0.63 0.64
1450395 at Slc22a5 solute carrier family 22 (organic cation transporter), member 5 3712 0.82 0.82
Figure imgf000064_0001
Figure imgf000064_0002
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000067_0002
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000070_0002
Figure imgf000071_0001
Figure imgf000073_0001
NCBA: normal CBA kidney; Iso CBA: CBA isograft in CBA host; AUo CBA-B6Nude: CBA allograft in B6 host; nB6: normal B6 kidney; nBalb/c: normal Balb/c kidney (wildtype); nBalb/c. GKO: normal GKO kidney (Balb/c background); CBA + rIFNK: kidney (CBA) from mouse treated with recombinant IFNK; B6 + rIFNK: kidney (B6) from mouse treated with recombinant IFNK; Balb/c+ rIFNK: kidney (Balb/c) from mouse treated with recombinant IFNK; Iso Balb/c: Balb/c isograft in Balb/c host; Iso Balb/cGKO: GKO isograft in GKO host, both on Balb/c background; AUo Balb/c-B6: Balb/c allograft in B6 host; AUo GKO-GKO: Balb/c.GKO allograft in B6.GKO host; AUo GRKO-B6: CBA.GRKO allograft in B6 host; AUo CBA-Nude: CBA allograft in B6 nude hosts; AUo Balb/c-B6: Balb/c allograft in B6 host; AUo CBA-GzmABKO: CBA allograft in Bό.GzmABKO host; AUo CBA-PrfKO: CBA allograft in Bό.PrfKO host; AUo CBA-IgKO: CBA allograft in Bό.IgKO host; AUo CBA-Balb/c: CBA allograft in Balb/c host; AUo CBA-CD103: CBA allograft in Balb/c.CD103 host; CBA ATN D7: ATN kidney (CBA); B6 ATN: ATN kidney (B6); Iso CBA host:
10 host kidney from CBA host with a CBA isograft; AUo CBA-B6 host: host kidney from B6 host with B6 allograft; Dilution: normal CBA kidney RNA diluted with RNA from MLR (4+1); MLR: mixed lymphocyte culture (CBA stimulators, B6 responder cells)
Table 2
Renal solute carrier transcripts decreased in allografts and isografts in response to transplant injury (humanized)
CO
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Table 3
Renal transcripts decreased in allografts and isografts with injury (mouse)
Figure imgf000078_0001
O
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
O
Figure imgf000083_0001
Figure imgf000084_0001
CO
Figure imgf000085_0001
O
Figure imgf000086_0001
O
Figure imgf000087_0001
O
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
CO
Figure imgf000099_0001
Figure imgf000100_0001
O O
Figure imgf000101_0001
O
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
O
Figure imgf000105_0001
O
Figure imgf000106_0001
Figure imgf000106_0002
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000109_0001
Figure imgf000110_0001
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
O
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000135_0002
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000163_0002
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Figure imgf000191_0001
Figure imgf000192_0001
Figure imgf000192_0002
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Table 4
Renal transcripts decreased in allografts and isografts with injury (humanized)
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
Figure imgf000235_0001
Table 5
NIRITs (mouse)
Figure imgf000236_0001
Figure imgf000237_0001
Figure imgf000238_0001
Figure imgf000239_0001
Figure imgf000240_0001
Figure imgf000241_0001
Figure imgf000242_0001
Figure imgf000243_0001
Figure imgf000246_0001
Figure imgf000247_0001
Figure imgf000248_0001
Figure imgf000249_0001
Figure imgf000250_0001
Figure imgf000251_0001
Figure imgf000252_0001
Table 6
NIRITs (humanized)
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
Figure imgf000258_0001
Figure imgf000259_0001
Figure imgf000260_0001
Figure imgf000261_0001
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
Table 7
Unique IRITs, IRIT-ATN, IRIT-host, IRIT-Dl, IRIT-D3 andIRIT-D5 (mouse)
Figure imgf000266_0001
Figure imgf000267_0001
Figure imgf000268_0001
CO
Figure imgf000269_0001
Figure imgf000270_0001
O
Figure imgf000271_0001
Figure imgf000272_0001
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
CO
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
O
Figure imgf000285_0001
O
Figure imgf000286_0001
CO
Figure imgf000287_0001
CO
Figure imgf000288_0001
Figure imgf000289_0001
CO
Figure imgf000290_0001
O
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0002
Figure imgf000297_0001
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0002
Figure imgf000311_0001
Figure imgf000312_0001
Figure imgf000313_0001
Figure imgf000314_0001
Table 8
IRITs (humanized)
Figure imgf000315_0001
Figure imgf000316_0001
Figure imgf000317_0001
Figure imgf000318_0001
Figure imgf000319_0001
Figure imgf000320_0001
Figure imgf000321_0001
Figure imgf000322_0001
Figure imgf000323_0001
Figure imgf000324_0001
Figure imgf000325_0001
Figure imgf000326_0001
Figure imgf000327_0001
Figure imgf000328_0001
Figure imgf000329_0001
Figure imgf000330_0001
Figure imgf000331_0001
Table 9
Macrophage associated transcripts (MATs) expressed in isografts - IRIT-MATs (mouse)
Figure imgf000332_0001
Figure imgf000333_0001
Figure imgf000334_0001
Figure imgf000335_0001
Figure imgf000336_0001
Figure imgf000337_0001
Figure imgf000338_0001
Figure imgf000339_0001
Figure imgf000340_0001
Figure imgf000341_0001
Table 10
Top 25 IRITs: Dl, D2, D3, D4, D5, D7, D21 (mouse)
Top 25 genes differentiating day 1 isografts from day 1 allografts iso D1 allo D1
VS VS corrected probe set ID Gene Symbol Gene Title control control allo/iso p value no significantly different genes
Top 25 genes differentiating day 2 isografts from < day 2 allografts iso D2 allo D2
VS VS corrected probe set ID Gene Symbol Gene Title control control allo/iso p value no significantly different genes
Genes differentiating day 3 isografts from day 3 allografts iso D3 allo D3
VS VS corrected probe set ID Gene Symbol Gene Title control control allo/iso p value
1460218_at Cd52 CD52 antigen 2.5 10.7 4.2 0.010 chemokine (C-C motif)
1421186_at Ccr2 receptor 2 1.8 6.5 3.6 0.010
1436996_x_at Lzp-s P lysozyme structural 5.8 16.2 2.8 0.010 lymphocyte antigen 6
1453304_s_at Ly6e complex, locus E 1.9 5.1 2.7 0.010
1448591 _at Ctss cathepsin S 3.6 9.2 2.5 0.014 transforming growth
1456250_x_at Tgfbi factor, beta induced 5.4 13.4 2.5 0.027
Fc receptor, IgE, high affinity I, gamma
1418340_at Fcerig polypeptide 2.9 7.1 2.5 0.010 lymphocyte cytosolic
1415983_at Lcp1 protein 1 2.6 6.3 2.4 0.014
TYRO protein tyrosine
1450792_at Tyrobp kinase binding protein 3.3 7.5 2.3 0.042
Rho, GDP dissociation
1426454_at Arhgdib inhibitor (GDI) beta 1.7 3.6 2.1 0.019 gremlin 2 homolog, cysteine knot superfamily (Xenopus
1418492 at Grem2 laevis) 2.3 1.0 0.5 0.023
Top 25 genes differentiating day 4 isografts from i day 4 allografts iso D4 allo D4
VS VS corrected
Probe set ID Gene Symbol Gene Title control control allo/iso p value
1460218_at Cd52 CD52 antigen 2.4 19.5 8.1 0.000
1438009_at MGC73635 similar to histone 2a 1.9 14.3 7.3 0.000 fibrinogen, alpha
1424279_at Fga polypeptide 0.8 5.2 6.5 0.000 lymphocyte cytosolic
1415983_at Lcp1 protein 1 1.8 11.2 6.3 0.000 1450788_at Saa1 serum amyloid A 1 1.1 5.4 5.1 0.000 chemokine (C-C motif)
1421186_at Ccr2 receptor 2 2.1 10.5 5.1 0.000 lymphocyte antigen 6
1453304_s_at Ly6e complex, locus E 2.0 8.9 4.4 0.000 1456292_a_at Vim vimentin 2.9 12.6 4.4 0.000 1448591 _at Ctss cathepsin S 3.6 14.4 4.0 0.000 transforming growth
1456250_x_at Tgfbi factor, beta induced 4.3 16.3 3.8 0.000
Fc receptor, IgE, high affinity I, gamma
1418340_at Fceii g polypeptide 2.6 9.7 3.7 0.000 complement
1423954_at C3 component 3 10.8 38.3 3.5 0.000 1436996_x_at Lzp-s P lysozyme structural 6.3 21.8 3.4 0.000 1417268_at Cd14 CD14 antigen 3.6 12.3 3.4 0.000
TYRO protein tyrosine
1450792_at Tyrobp kinase binding protein 3.5 11.8 3.4 0.000
Rho, GDP dissociation
1426454_at Arhgdib inhibitor (GDI) beta 1.5 4.9 3.3 0.000
PYD and CARD
1417346_at Pycard domain containing 1.8 6.0 3.3 0.000 nucleolar and spindle
1416309_at Nusapi associated protein 1 1.3 4.2 3.2 0.001 1437185_s_at TmsbiO thymosin, beta 10 2.4 7.0 3.0 0.001 procollagen, type III,
1427884_at Col3a1 alpha 1 3.4 10.1 3.0 0.001 aldehyde dehydrogenase family
1422789_at Aldh1a2 1 , subfamily A2 3.0 8.9 3.0 0.002 intercellular adhesion
1424067_at Icami molecule 2.8 8.2 2.9 0.001 procollagen, type I,
1423669 at CoH a1 alpha 1 3.2 9.0 2.8 0.001
SH3 domain binding glutamic acid-rich
1416528 at Sh3bgrl3 protein-like 3 1.6 4.4 2.8 0.002
Top 25 genes differentiating day 5 isografts from day 5 allografts iso D5 allo D5 vs vs corrected
Probe set ID Gene Symbol Gene Title control control allo/iso p value
1460218_at Cd52 CD52 antigen 4.2 31.2 7.3 0.006
1450788_at Saa1 serum amyloid A 1 1.2 8.3 6.9 0.041 lymphocyte cytosolic
1415983_at Lcp1 protein 1 2.9 17.0 5.8 0.010
1438009_at MGC73635 similar to histone 2a 4.8 27.2 5.7 0.010 transforming growth
1456250 x at Tgfbi factor, beta induced 6.0 32.4 5.4 0.010 chemokine (C-C motif)
1421186_at Ccr2 receptor 2 3.6 18.7 5.1 0.015
Rho, GDP dissociation
1426454_at Arhgdib inhibitor (GDI) beta 2.0 8.6 4.4 0.010
PYD and CARD
1417346_at Pycard domain containing 2.1 7.9 3.8 0.011 lymphocyte antigen 6
1453304_s_at Ly6e complex, locus E 2.8 10.1 3.6 0.015 1456292_a_at Vim vimentin 5.0 17.7 3.5 0.041
Fc receptor, IgE1 high affinity I, gamma
1418340_at Fceri g polypeptide 4.7 16.2 3.5 0.027
TYRO protein tyrosine
1450792_at Tyrobp kinase binding protein 5.3 18.2 3.4 0.029
FXYD domain- containing ion
1418296_at Fxydδ transport regulator 5 2.8 9.4 3.3 0.027 1426501 _a_at T2bp Traf2 binding protein 1.6 5.2 3.2 0.027 1448591 _at Ctss cathepsin S 6.4 20.2 3.2 0.027 ribonucleotide
1448226_at Rrm2 reductase M2 2.7 8.4 3.1 0.043 kinesin family member
1435306_a_at Kif11 11 1.5 4.6 3.1 0.029 nucleolar and spindle
1416309_at Nusapi associated protein 1 2.5 6.9 2.8 0.037 1429399_at Rnf125 ring finger protein 125 1.6 4.3 2.7 0.041 1434248_at Prkch protein kinase C, eta 1.3 3.5 2.6 0.027
Friend leukemia
1433512_at RIiI integration 1 3.0 7.0 2.4 0.041 procollagen, type XIV,
1428455_at Col14a1 alpha 1 1.8 0.8 0.4 0.041 solute carrier family 14
(urea transporter),
1425250_a_at SId 4a2 member 2 1.7 0.7 0.4 0.041
DNA segment, human
1450839 at D0H4S114 D4S114 1.5 0.5 0.3 0.029
Top 25 genes differentiating day 7 isografts from day 7 allografts iso D7 allo D7 vs vs corrected
Probe set ID Gene Symbol Gene Title control control allo/iso p value
1460218_at Cd52 CD52 antigen 3.7 40.9 10.9 0.000 lymphocyte antigen 6
1453304_s_at Ly6e complex, locus E 1.8 16.0 8.8 0.000 lymphocyte cytosolic 1415983_at Lcp1 protein 1 2.7 22.9 8.4 0.000 transforming growth
1456250_x_at Tgfbi factor, beta induced 5.5 44.3 8.1 0.000 chemokine (C-C motif)
1421186_at Ccr2 receptor 2 3.7 29.6 8.0 0.000
Fc receptor, IgE, high
1418340_at Fceri g affinity I, gamma 4.1 28.5 7.0 0.000 polypeptide
FXYD domain- containing ion
1418296_at Fxydδ transport regulator 5 2.4 16.9 6.9 0.000
1456292_a_at Vim vimentin 3.9 26.8 6.9 0.000
TYRO protein tyrosine
1450792_at Tyrobp kinase binding protein 4.8 31.9 6.7 0.000
Rho, GDP dissociation
1426454_at Arhgdib inhibitor (GDI) beta 1.7 11.4 6.6 0.000 procollagen, type I,
1423669_at CoH a1 alpha 1 4.4 27.7 6.3 0.000 colony stimulating factor 2 receptor, alpha, low-affinity
(granulocyte-
1420703_at Csf2ra macrophage) 1.7 9.6 5.7 0.000
1438009_at MGC73635 similar to histone 2a 4.2 23.2 5.6 0.000 microfibrillar
1418454_at Mfapδ associated protein 5 3.3 18.5 5.5 0.000 actin related protein
2/3 complex, subunit
1416226_at Arpd b 1 B 3.4 18.0 5.4 0.000
PYD and CARD
1417346_at Pycard domain containing 2.3 11.7 5.1 0.000
1448591 _at Ctss cathepsin S 6.0 30.5 5.1 0.000
1434248_at Prkch protein kinase C, eta 1.2 5.8 4.8 0.000 procollagen, type V,
1422437_at Col5a2 alpha 2 4.5 20.6 4.6 0.000
1438200_at SuIH sulfatase 1 1.7 7.9 4.6 0.000
SH3 domain binding glutamic acid-rich
1416528_at Sh3bgrl3 protein-like 3 1.8 8.1 4.6 0.000 cytokine receptor-like
1418476_at CrIfI factor 1 2.0 9.2 4.5 0.000 procollagen, type V,
1416740_at Col5a1 alpha 1 1.9 8.5 4.5 0.000 complement component 1, q subcomponent, alpha
1417381_at C1qa polypeptide 6.7 29.2 4.3 0.000 intercellular adhesion
1424067 at Icami molecule 2.6 11.3 4.3 0.000
Top 25 genes differentiating day 21 isografts from day 21 allografts iso allo
D21 vs D21 vs corrected
Probe set ID Gene Symbol Gene Title control control allo/iso p value
1460218_at Cd52 CD52 antigen 2.1 36.1 17.5 0.000 microfibrillar
1418454_at Mfap5 associated protein 5 2.2 34.4 15.8 0.000 complement component 1 , q subcomponent, alpha
1417381 _at C1qa polypeptide 2.7 39.9 14.6 0.000
Fc receptor, IgE, high affinity I, gamma
1418340_at Fceri g polypeptide 2.0 27.8 14.0 0.000
TYRO protein tyrosine
1450792_at Tyrobp kinase binding protein 2.2 31.2 13.9 0.000
1438009_at MGC73635 similar to histone 2a 0.5 6.7 13.9 0.000
1456292_a_at Vim vimentin 1.8 21.5 11.8 0.000
1448591 _at Ctss cathepsin S 2.8 32.4 11.5 0.000
EGF-like module containing, mucin-like, hormone receptor-like
1451161 _a_at EmM sequence 1 1.5 17.3 11.4 0.000 complement
1423954_at C3 component 3 3.6 40.2 11.2 0.000 procollagen, type III,
1427884_at Col3a1 alpha 1 3.1 33.1 10.7 0.000 transforming growth
1456250_x_at Tgfbi factor, beta induced 2.6 27.3 10.6 0.000 procollagen, type V,
1422437_at Col5a2 alpha 2 2.1 19.8 9.5 0.000 lymphocyte cytosolic
1415983_at Lcp1 protein 1 2.0 19.2 9.5 0.000 lymphocyte antigen 6
1453304_s_at Ly6e complex, locus E 1.7 15.1 9.1 0.000 chemokine (C-C motif)
1421186_at Ccr2 receptor 2 1.8 15.4 8.5 0.000
1422571 _at Thbs2 thrombospondin 2 1.4 11.6 8.1 0.000 cytokine receptor-like
1418476_at CrIfI factor 1 0.9 7.4 8.0 0.000
PYD and CARD
1417346_at Pycard domain containing 1.1 8.9 8.0 0.000 colony stimulating factor 2 receptor, alpha, low-affinity
(granulocyte-
1420703_at Csf2ra macrophage) 1.2 9.4 7.9 0.000 procollagen, type I,
1423110_at CoI 1a2 alpha 2 2.0 14.9 7.5 0.000 serine (or cysteine) peptidase inhibitor,
1422804_at Serpinbβb clade B, member 6b 1.2 8.4 7.2 0.000
1426642_at Fn1 fibronectin 1 1.7 11.9 7.0 0.000 ribonucleotide
1448226 at Rrm2 reductase M2 0.8 5.8 7.0 0.000 Table 11
Full list of unique GSTs, showing fold increase in IFN-K deficient isografts (BALB.GKO) and allografts (BALB.GKO, CBA.GRKO) compared to WT (BALB, CBA) grafts at day 5 or day 7, and in WT CBA allografts at day 42 compared to control kidneys (NCBA)
(mouse)
Figure imgf000347_0001
Figure imgf000348_0001
Figure imgf000349_0001
Figure imgf000350_0001
Figure imgf000351_0001
Figure imgf000352_0001
Figure imgf000353_0001
Figure imgf000354_0001
Figure imgf000355_0001
Figure imgf000356_0001
Figure imgf000357_0001
Figure imgf000358_0001
Figure imgf000359_0001
Figure imgf000360_0001
Figure imgf000361_0001
GST are listed in the alphabetical order. BALB. GKO isografts were compared to WT BALB isografts. BALB. GKO allografts were compared to the WT (BALB) allografts, CBA.GRKO allografts were compared to the WT CBA allografts and WT CBA allografts
day 42 were compared to control kidneys (NCBA). All allografts were from the B6 hosts. Bolded numbers indicate significant expression of GSTs (FDR=0.05) in CBA allografts d42. Function column lists the GO categories and flags the AMA markers.
CO <7>
Table 12
Full list of unique GSTs, showing fold increase in IFN-K deficient isografts (B ALB. GKO) and allografts (BALB. GKO, CBA. GRKO) compared to WT (BALB, CBA) grafts at day 5 or day 7, and in WT CBA allografts at day 42 compared to control kidneys (NCBA)
(humanized)
Figure imgf000363_0001
Figure imgf000364_0001
Figure imgf000365_0001
Table 13
Unique CISTs (mouse)
Figure imgf000366_0001
Figure imgf000367_0001
Figure imgf000368_0001
Figure imgf000369_0001
Unique genes are listed in descending order by their fold change in Tap IKO day 7 vs B6 day 7.
Table 14
CISTs (humanized)
Figure imgf000370_0001
Figure imgf000371_0001
Table 15
Cytotoxic T cell associated transcripts (CATs - mouse)
Figure imgf000372_0001
Figure imgf000373_0001
Figure imgf000374_0001
Figure imgf000375_0001
Figure imgf000376_0001
Figure imgf000377_0001
Figure imgf000378_0001
Figure imgf000379_0001
Table 16 tGRITs
Figure imgf000380_0001
Figure imgf000381_0001
Table 17
Characteristics of the DD kidneys0 in the 'Low Risk' and 'High Risk' clusters
Figure imgf000382_0001
26 kidneys from 18 male donors and 16 from 13 female donors; dumber of measurements 2global kidney score (JASN 1999) 3intra-operative mean arterial pressure
4fall of serum creatinine from day 1 to day 2, not calculated in case of renal replacement therapy on day 1
Excluding creatinine measurements during ongoing renal replacement therapy 6Cockroft-Gault formula: ((140 - R age) * R lean body weight * R gender) / (72 * R crea * 0.0113)
7cytomegalo virus (CMV) disease was diagnosed in case of viremia plus clinical symptoms/signs plus treatment Table 18
Risk factors and delayed graft function rates in the DD kidneys0 in the 'Low Risk' and
'Hi h Risk' clusters
Figure imgf000383_0001
26 kidneys from 18 male donors and 16 from 13 female donors
1 3 patients in cluster 2 have missing values
2 1 patient in cluster 3 has missing value Table 19
Genes that correlate with Slcs (top 25 that positively correlate and top 25 that negatively correlate)
Negatively correlated with Slcs Positively correlated with Slcs
LOC162073 -0.86848 PEPD 0.943178
CBFB -0.85556 BHMT2 0.937548
ANXA2 -0.84999 AGMAT 0.929208
ECOP -0.84684 MSRA 0.925029
RCN 1 -0.84608 ALDH2 0.923828
NOTCH2 -0.84471 KHK 0.919334
ABCC1 -0.84454 GALM 0.91535
CD47 -0.8424 DPYS 0.913884
AHNAK -0.84223 PC 0.913581
LOC253981 -0.84182 LOC134147 0.910261
NSMAF -0.84052 UPB1 0.909611
RAB27A -0.8379 ALDH6A1 0.908844
RPL39 -0.83514 SORD 0.908648
KBTBD2 -0.83507 SHMT1 0.908529
RBM17 -0.83487 AGPAT3 0.907917
LNPEP -0.83424 ALDOB 0.905693
NAP1 L1 -0.82972 SLC12A6** 0.90431
IL13RA1 -0.82947 DHTKD1 0.90225
UBE2E1 -0.82922 ACOX2 0.901203
FUT11 -0.82889 GPD1 0.900891
WTAP -0.82858 BHMT 0.900222
SEPT8 -0.82704 ABHD6 0.899886
S100A10 -0.82693 XPNPEP2 0.898105
ANKRD13A -0.82451 C7orf10 0.896816
FER1 L3 -0.8241 FLJ20920 0.896266
Table 20
Genes that correlate with IRITs (top 25 that positively correlate and top 25 that negatively correlate)
Negatively correlated with IRITs Positively correlated with IRITs
ACAT1 -0.85048 C1 R 0.904293
PANK1 -0.83548 TIMP1 0.902533
CYB5A -0.8351 LOXL1 0.865647
AK3L1 -0.83121 C1S 0.864847
PECI -0.81544 C9orf19 0.861084
ABHD10 -0.81536 PXDN 0.858749
GLYAT -0.81131 CKAP4 0.858198
HSPD1 -0.81 TPBG 0.856504
AFTPH -0.80999 SLC43A3 0.852626
ACADSB -0.80984 ACTG 1 0.849592
ALDH6A1 -0.80903 LOC340061 0.849096
ASPA -0.80887 EMILIN2 0.846306
FLJ38482 -0.80816 TBC1D2B 0.845222
METTL7A -0.80778 SHC1 0.843507
ACADM -0.80642 CSPG2 0.841406
TRPM3 -0.80604 PLXNA1 0.839986
TRIM10 -0.80603 ELF4 0.838451
GBA3 -0.80225 C22orf9 0.836235
HAO2 -0.802 FLJ38984 0.831815
GSTA3 -0.79744 SERPING1 0.830855
BHMT -0.79606 CD44 0.829841
GSTA2 -0.79543 AYTL2 0.829443
DMGDH -0.79477 ECOP 0.825823
CLYBL -0.79163 IFITM2 0.824929
PRLR -0.79138 NECAP2 0.823894
Table 21
Pathways that correlate with Slcs (top 50 pathways)
Top 25 pathways that negatively correlate Top 25 pathways that positively correlate with Slcs with Slcs
Glyoxylate and dicarboxylate metabolism Inositol metabolism
Pathogenic Escherichia coli infection EHEC Benzoate degradation via hydroxylation
Pathogenic Escherichia coli infection EPEC Styrene degradation
Nitrogen metabolism Vitamin B6 metabolism
Metabolism of xenobiotics by cytochrome P450 Fatty acid elongation in mitochondria
Ribosome beta Alanine metabolism
Limonene and pinene degradation Valine leucine and isoleucine degradation
Benzoate degradation via CoA ligation Glyoxylate and dicarboxylate metabolism
Atrazine degradation Propanoate metabolism
Cell Communication Fatty acid metabolism
D Glutamine and D glutamate metabolism Cyanoamino acid metabolism
Bile acid biosynthesis Glycine serine and threonine metabolism
Chondroitin sulfate biosynthesis Ascorbate and aldarate metabolism
Cholera Infection Alanine and aspartate metabolism
Dorso ventral axis formation Pyruvate metabolism
Heparan sulfate biosynthesis Carbon fixation gamma Hexachlorocyclohexane degradation Arginine and proline metabolism
Cell cycle Urea cycle and metabolism of amino groups
Ethylbenzene degradation Alkaloid biosynthesis I
Glutathione metabolism Citrate cycle TCA cycle
Pantothenate and CoA biosynthesis Methane metabolism
ECM receptor interaction Bile acid biosynthesis
Arachidonic acid metabolism Histidine metabolism
Alkaloid biosynthesis Il Pentose phosphate pathway
Phenylalanine metabolism Glycolysis Gluconeogenesis
Table 22
Pathways that correlate with IRITs (top 50 pathways)
Top 25 pathways that negatively correlate Top 25 pathways that positively correlate with IRITs with IRITs
Inositol metabolism Glutathione metabolism
Styrene degradation Fatty acid elongation in mitochondria
Glyoxylate and dicarboxylate metabolism Cell Communication
Benzoate degradation via hydroxylation Atrazine degradation
Valine leucine and isoleucine degradation Propanoate metabolism
Fatty acid elongation in mitochondria Pathogenic Escherichia coli infection EHEC beta Alanine metabolism Pathogenic Escherichia coli infection EPEC
Propanoate metabolism Chondroitin sulfate biosynthesis
Fatty acid metabolism ECM receptor interaction
Citrate cycle TCA cycle Metabolism of xenobiotics by cytochrome P450
Vitamin B6 metabolism Pentose and glucuronate interconversions
Bile acid biosynthesis Nitrogen metabolism
Ascorbate and aldarate metabolism Urea cycle and metabolism of amino groups
Alkaloid biosynthesis I Cholera Infection
Ubiquinone biosynthesis beta Alanine metabolism
Carbon fixation Leukocyte transendothelial migration
Reductive carboxylate cycle CO2 fixation Bile acid biosynthesis
Pentose phosphate pathway Glyoxylate and dicarboxylate metabolism
Glycine serine and threonine metabolism Focal adhesion
Stilbene coumarine and lignin biosynthesis Glycosaminoglycan degradation
Histidine metabolism Glycolysis Gluconeogenesis
Alanine and aspartate metabolism X24 Dichlorobenzoate degradation
Arginine and proline metabolism Alanine and aspartate metabolism
Pyruvate metabolism Ribosome
Lysine biosynthesis Carbon fixation
Table 23
Figure imgf000388_0001
Interstitial infiltrate, graft necrosis, edema and peritubular capillary congestion (PTC) were recorded as a percentage positive of the whole cortex area. Tubulitis was scored as the number of tubules with tubulitis in one tissue cross section (for NCBA, Iso D5, Iso D7, Iso D21, WT D3, WT D4, WT D5, WT D7) or in ten high power fields (WT D14, WT D21, WT D42). Arteritis and venulitis lesions were counted and given as the mean number of involved vessels per kidney section. The numbers shown are mean ± standard deviation.
Table 24
Histopathologic changes in CBA into wild-type BALB/c (WT, n=5) and in CBA into 103-/- BALB/c (CD 103-/-, n=4) at day 21 post transplant. There were no significant differences between WT and CD 103-/- other than for edema (p < 0.05).
WT CD103"'"
Histology1
(n = 5) (n = 4)
Weight (mg) 340 ± 123 338 ± 48
Necrosis (%) 4.0 ± 8.9 23 ± 21
PTC (%) 20 ± 24 53 ± 25
Glomerulitis 2.8 ± 0.4 3.0 ± 0.0
Tubulitis (%) 64 ± 5.5 63 ± 10
Int Infiltrate (%) 56 ± 5.5 43 ± 10
Arteritis 1.2 ± 1.1 1.8 ± 1.0
Art Thrombosis 0.0 0.0
Venulitis 1.8 ± 1.3 1.8 ± 1.7
Ven Thrombosis 6.2 ± 8.6 0.0
Edema (%) 6.0 ± 8.9 23 ± 3
Cast 0.0 0.0
Interstitial infiltrate, tubulitis, graft necrosis, edema, and peritubular capillary congestion (PTC) were recorded as a percentage positive of the whole cortex area. Glomerulitis lesions were scored from 0 to 3 (0 = no change, 1 = 0-25%, 2 = 25- 75%, and 3 = 75-100% of the total parenchyma involved). Arteritis and venulitis lesions were counted and given as the mean number of involved vessels per kidney section. The numbers shown are mean values ± standard deviation.
Table 25
Expression of glucose transporters as assessed by microarray in isografts (Iso), allografts rejecting in wildtype B6 hosts (WT), contralateral host kidney (Left), kidneys with ischemic acute tubular necrosis (ATN), cultured cytotoxic lymphocytes (CTL), and mixed lymphocyte culture (MLR). Only those glucose transporters that were present in NCBA and had low expression in CTL are represented in this table. Numbers represent signal strength for NCBA and fold changes compared to NCBA for all other experimental groups. A)
MOE 430A array and B) MOE 430 2.0 array.
Figure imgf000390_0001
Table 26
Expression of amino acid transporters as assessed by microarray in isografts (Iso), allografts rejecting in wildtype B6 hosts (WT), contralateral host kidney (Left), kidneys with ischemic acute tubular necrosis (ATN), cultured cytotoxic lymphocytes (CTL), and mixed lymphocyte culture (MLR).
Only those amino acid transporters that were present in NCBA and had low expression in CTL are represented in this table. Numbers represent signal strength for NCBA and fold changes compared to NCBA for all other experimental groups. A) MOE 430A array and B) MOE 4302.0 array.
Figure imgf000391_0001
Table 27
Expression ofaquaporins as assessed by microarray in isografts (Iso), allografts rejecting in wildtype B6 hosts (WT), contralateral host kidney (Left), kidneys with ischemic acute tubular necrosis (ATN), cultured cytotoxic lymphocytes (CTL), and mixed lymphocyte culture
(MLR). Only those aquaporins that were present in NCBA and had low expression in CTL are represented in this table. Numbers represent signal strength for NCBA and fold changes compared to NCBA for all other experimental groups. A) MOE 430A array and B)
MOE 430 2.0 array.
Figure imgf000392_0001
Table 28
Expression of glucose transporters, amino acid transporters, and aquaporins in allografts rejecting in wild-type Balb/c hosts (WT) and in CDlOS-deficient hosts (CD 103'
'') at day 21 post transplant, as assessed by microarray.
Figure imgf000393_0001
Numbers represent signal strength for normal CBA kidney (NCBA) and fold changes compared to NCBA for rejecting allografts.
Table 29
Transcript expression of glucose transporters, amino acid transporters, and aquaporins in allografts in hosts lacking perform (Prf '), granzyme A and gran∑yme B (GzmAB' ') or mature B-cells (Ig' '). Numbers represent signal strength for NCBA and fold changes compared to NCBA for all other experimental group. GzmABKO and PrfKO were analyzed on MOE4302.0, IghKO on
MOE430Aarrays.
GzmAB PrfΛ GzmAB " Prf " is "
Transcript NCBA D7 D7 D21 D21 NCBA D7 D21
Glucose Transporters
Slc2a2 9441 0.08 0.18 0.13 0.2 3192 0.18 0.39
Slc2a4 671 0.18 0.34 0.17 0.17 552 0.44 0.51
Slc2a5 3904 0.08 0.18 0.07 0.05 1778 0.26 0.35
Slc5al 1789 0.55 0.87 0.57 0.9 779 0.69 0.85
Slc5alO 1180 0.07 0.19 0.04 0.14 572 0.27 0.53
Slc5a2 6280 0.01 0.04 0.02 0.01 2397 0.07 0.27
Amino Acid Transporters
Slclal 6505 0.27 0.34 0.15 0.16 2511 0.5 0.49
Slcla4 551 0.96 1.32 0.62 0.52 257 1.55 0.98
Slc3al 13224 0.15 0.29 0.12 0.14 5852 0.29 0.42
Slc6al3 1612 0.1 0.28 0.1 0.12 794 0.38 0.39
Slc7alO 7889 0.22 0.32 0.21 0.24 4001 0.38 0.56
Slc7al3 1301 0.02 0.1 0.05 0.01 364 0.17 0.57
Slc7al3 25518 0.02 0.14 0.07 0.02 4345 0.14 0.86
Slc7a8 730 0.34 0.37 0.34 0.37 639 0.24 0.34
Slc7a9 10876 0.1 0.2 0.11 0.15 5408 0.31 0.45
Figure imgf000395_0001
OTHER EMBODIMENTS
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A method for detecting tissue injury, wherein said method comprises determining whether or not a tissue contains cells having an injury and repair profile, wherein the presence of said cells indicates that said tissue is injured.
2. The method of claim 1, wherein said mammal is a human.
3. The method of claim 1 , wherein said tissue is from a biopsy.
4. The method of claim 1 , wherein said tissue is kidney tissue.
5. The method of claim 1, wherein said tissue is to be transplanted into a recipient.
6. The method of claim 1 , wherein said tissue has been transplanted into a recipient.
7. The method of claim 1, wherein said determining step comprises using PCR or a nucleic acid array.
8. The method of claim 1, wherein said determining step comprises using immunohistochemistry or an array for detecting polypeptides.
9. A method for detecting tissue injury, wherein said method comprises determining whether or not a tissue contains cells having a not-in-isografts injury and repair profile, wherein the presence of said cells indicates that said tissue is injured.
10. The method of claim 9, wherein said mammal is a human.
11. The method of claim 9, wherein said tissue is from a biopsy.
12. The method of claim 9, wherein said tissue is kidney tissue.
13. The method of claim 9, wherein said tissue is to be transplanted into a recipient.
14. The method of claim 9, wherein said tissue has been transplanted into a recipient.
15. The method of claim 9, wherein said determining step comprises using PCR or a nucleic acid array.
16. The method of claim 9, wherein said determining step comprises using immunohistochemistry or an array for detecting polypeptides.
17. A method for detecting tissue injury, wherein said method comprises determining whether or not a tissue contains cells having a gamma interferon (IFN-K) suppressed profile, wherein the presence of said cells indicates that said tissue is injured.
18. The method of claim 17, wherein said mammal is a human.
19. The method of claim 17, wherein said tissue is from a biopsy.
20. The method of claim 17, wherein said tissue is kidney tissue.
21. The method of claim 17, wherein said tissue is to be transplanted into a recipient.
22. The method of claim 17, wherein said tissue has been transplanted into a recipient.
23. The method of claim 17, wherein said determining step comprises using PCR or a nucleic acid array.
24. The method of claim 17, wherein said determining step comprises using immunohistochemistry or an array for detecting polypeptides.
25. A method for detecting tissue injury, wherein said method comprises determining whether or not a tissue contains cells having a class I suppressed profile, wherein the presence of said cells indicates that said tissue is injured.
26. The method of claim 25, wherein said mammal is a human.
27. The method of claim 25, wherein said tissue is from a biopsy.
28. The method of claim 25, wherein said tissue is kidney tissue.
29. The method of claim 25, wherein said tissue is to be transplanted into a recipient.
30. The method of claim 25, wherein said tissue has been transplanted into a recipient.
31. The method of claim 25, wherein said determining step comprises using PCR or a nucleic acid array.
32. The method of claim 25, wherein said determining step comprises using immunohistochemistry or an array for detecting polypeptides.
33. A method for detecting tissue injury, wherein said method comprises determining whether or not a tissue contains cells having a renal transcript (RT) profile, wherein the presence of said cells indicates that said tissue is injured.
34. The method of claim 33, wherein said mammal is a human.
35. The method of claim 33, wherein said tissue is from a biopsy.
36. The method of claim 33, wherein said tissue is kidney tissue.
37. The method of claim 33, wherein said tissue is to be transplanted into a recipient.
38. The method of claim 33, wherein said tissue has been transplanted into a recipient.
39. The method of claim 33, wherein said determining step comprises using PCR or a nucleic acid array.
40. The method of claim 33, wherein said determining step comprises using immunohistochemistry or an array for detecting polypeptides.
41. A method for detecting tissue injury, wherein said method comprises determining whether or not a tissue contains cells having a solute carrier profile, wherein the presence of said cells indicates that said tissue is injured.
42. The method of claim 41, wherein said mammal is a human.
43. The method of claim 41 , wherein said tissue is from a biopsy.
44. The method of claim 41 , wherein said tissue is kidney tissue.
45. The method of claim 41, wherein said tissue is to be transplanted into a recipient.
46. The method of claim 41, wherein said tissue has been transplanted into a recipient.
47. The method of claim 41, wherein said determining step comprises using PCR or a nucleic acid array.
48. The method of claim 41, wherein said determining step comprises using immunohistochemistry or an array for detecting polypeptides.
49. A method for assessing whether a tissue is at risk for delayed graft function (DGF), wherein said method comprises determining whether or not a tissue contains cells having an injury and repair profile, a non-in-isografts injury and repair profile, an IFN-K suppressed profile, a class I suppressed profile, a renal transcript (RT) profile, or a solute carrier (SIc) profile, wherein the presence of said cells indicates that said tissue is at risk for DGF.
50. The method of claim 49, wherein said mammal is a human.
51. The method of claim 49, wherein said tissue is from a biopsy.
52. The method of claim 49, wherein said tissue is kidney tissue.
53. The method of claim 49, wherein said tissue is to be transplanted into a recipient.
54. The method of claim 49, wherein said tissue has been transplanted into a recipient.
55. The method of claim 49, wherein said determining step comprises using PCR or a nucleic acid array.
56. The method of claim 49, wherein said determining step comprises using immunohistochemistry or an array for detecting polypeptides.
57. A method for predicting whether a transplanted tissue will recover from injury, wherein said method comprises determining whether or not a tissue contains cells having an injury and repair profile, a non-in-isografts injury and repair profile, an IFN-K suppressed profile, a class I suppressed profile, a RT profile, or a SIc profile, wherein the presence of said cells indicates that said tissue is not likely to recover from injury.
58. The method of claim 57, wherein said mammal is a human.
59. The method of claim 57, wherein said tissue is from a biopsy.
60. The method of claim 57, wherein said tissue is kidney tissue.
61. The method of claim 57, wherein said tissue is to be transplanted into a recipient.
62. The method of claim 57, wherein said tissue has been transplanted into a recipient.
63. The method of claim 57, wherein said determining step comprises using PCR or a nucleic acid array.
64. The method of claim 57, wherein said determining step comprises using immunohistochemistry or an array for detecting polypeptides.
65. A method for detecting tissue injury, wherein said method comprises determining whether or not a tissue contains cells having an injury and repair correlated profile or an SIc correlated profile, wherein the presence of said cells indicates that said tissue is injured.
66. The method of claim 65, wherein said mammal is a human.
67. The method of claim 65, wherein said tissue is from a biopsy.
68. The method of claim 65, wherein said tissue is kidney tissue.
69. The method of claim 65, wherein said tissue is to be transplanted into a recipient.
70. The method of claim 65, wherein said tissue has been transplanted into a recipient.
71. The method of claim 65, wherein said determining step comprises using PCR or a nucleic acid array.
72. The method of claim 65, wherein said determining step comprises using immunohistochemistry or an array for detecting polypeptides.
73. A method for detecting tissue injury, comprising determining whether or not a tissue contains cells having increased activity of biochemical pathways that correlate with an injury and repair profile, with an SIc profile, with a non-in-isografts injury and repair profile, with a gamma interferon suppressed profile, with a class I suppressed profile, or with an RT profile, wherein the presence of said cells indicates that said tissue is injured.
74. The method of claim 73, wherein said biochemical pathways correlate with an injury and repair profile.
75. The method of claim 73, wherein said biochemical pathways correlate with an SIc profile.
76. A nucleic acid array comprising at least 20 nucleic acid molecules, wherein each of said at least 20 nucleic acid molecules has a different nucleic acid sequence, and wherein at least 50 percent of the nucleic acid molecules of said array comprise a sequence from nucleic acid selected from the group consisting of the nucleic acids listed in Tables 1-14, 19, and 20.
77. The array of claim 76, wherein said array comprises at least 50 nucleic acid molecules, wherein each of said at least 50 nucleic acid molecules has a different nucleic acid sequence.
78. The array of claim 76, wherein said array comprises at least 100 nucleic acid molecules, wherein each of said at least 100 nucleic acid molecules has a different nucleic acid sequence.
79. The array of claim 76, wherein each of said nucleic acid molecules that comprise a sequence from nucleic acid selected from said group comprises no more than three mismatches.
80. The array of claim 76, wherein at least 75 percent of the nucleic acid molecules of said array comprise a sequence from nucleic acid selected from said group.
81. The array of claim 76, wherein at least 95 percent of the nucleic acid molecules of said array comprise a sequence from nucleic acid selected from said group.
82. The array of claim 76, wherein said array comprises glass.
83. The array of claim 76, wherein said at least 20 nucleic acid molecules comprise a sequence present in a human.
84. A computer-readable storage medium having instructions stored thereon for causing a programmable processor to determine whether one or more nucleic acids listed in Tables 5-14, and the third column of Table 20 are present in a tissue sample at elevated levels.
85. The computer-readable storage medium of claim 84, further comprising instructions stored thereon for causing a programmable processor to determine whether one or more of the nucleic acids listed in Tables 5-14, and the third column of 20 is expressed at a greater level in said tissue sample than in a control tissue sample.
86. A computer-readable storage medium having instructions stored thereon for causing a programmable processor to determine whether one or more nucleic acids listed in Tables 1-4 and the third column of Table 19 are present in a tissue sample at decreased levels.
87. The computer-readable storage medium of claim 86, further comprising instructions stored thereon for causing a programmable processor to determine whether one or more of the nucleic acids listed in Tables 1-4 and the third column of Table 19 is expressed at a lower level in said tissue sample than in a control tissue sample.
88. An apparatus for determining whether a tissue is injured, said apparatus comprising: one or more collectors for obtaining signals representative of the presence of one or more nucleic acids listed in Tables 1-14, 19, and 20 in a sample from said tissue; and a processor for analyzing said signals and determining whether said tissue is injured.
89. The apparatus of claim 88, wherein said one or more collectors are configured to obtain further signals representative of the presence of said one or more nucleic acids in a control sample.
90. A method for detecting tissue rejection, wherein said method comprises determining whether or not tissue transplanted into a mammal contains cells that express a reduced level of a cadherin polypeptide or a transporter polypeptide, wherein the presence of said cells indicates that said tissue is being rejected.
91. The method of claim 90, wherein said mammal is a human.
92. The method of claim 90, wherein said tissue is kidney tissue.
93. The method of claim 90, wherein said tissue is a kidney.
94. The method of claim 90, wherein said method comprises determining whether or not said tissue contains cells that express a reduced level of said cadherin polypeptide.
95. The method of claim 90, wherein said cadherin polypeptide is an E-cadherin polypeptide or a Ksp-cadherin polypeptide.
96. The method of claim 90, wherein said method comprises determining whether or not said tissue contains cells that express a reduced level of said transporter polypeptide.
97. The method of claim 90, wherein said transporter polypeptide is selected from the group consisting of Slc2a2, Slc2a4, Slc2a5 Slc5al, Slc5a2, Slc5alO, Slc7a7, Slc7a8, Slc7a9, Slc7alO, Slc7al2, Slc7al3, Slcla4, Slc3al, Slclal, aquaporin 1, aquaporin 2, aquaporin 3, aquaporin 4, ABC transporter, solute carrier, and ATPase polypeptides.
98. The method of claim 90, wherein said determining step comprises measuring the level of mRNA encoding said cadherin polypeptide or said transporter polypeptide.
99. The method of claim 90, wherein said determining step comprises measuring the level of said cadherin polypeptide or said transporter polypeptide.
100. The method of claim 90, wherein said method comprises determining whether or not said tissue contains cells that express said cadherin polypeptide or said transporter polypeptide at a level less than the average level of expression exhibited in cells from control tissue that has not been transplanted.
101. The method of claim 90, wherein said determining step comprises determining whether or not a sample contains said cells, wherein said sample comprises cells, was obtained from tissue that was transplanted into said mammal, and was obtained from said tissue within fifteen days of said tissue being transplanted into said mammal.
PCT/CA2007/001295 2006-07-21 2007-07-20 Tissue rejection WO2008009132A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/374,639 US20090176656A1 (en) 2006-07-21 2007-07-20 Tissue rejection
EP07784962A EP2049713A4 (en) 2006-07-21 2007-07-20 Tissue rejection

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US82007306P 2006-07-21 2006-07-21
US60/820,073 2006-07-21
US92395407P 2007-04-17 2007-04-17
US60/923,954 2007-04-17

Publications (1)

Publication Number Publication Date
WO2008009132A1 true WO2008009132A1 (en) 2008-01-24

Family

ID=38956488

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2007/001295 WO2008009132A1 (en) 2006-07-21 2007-07-20 Tissue rejection

Country Status (3)

Country Link
US (1) US20090176656A1 (en)
EP (1) EP2049713A4 (en)
WO (1) WO2008009132A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010096160A1 (en) * 2009-02-17 2010-08-26 The Board Of Trustees Of The Leland Stanford Junior University Compartment-specific non-hla targets for diagnosis and prediction of graft outcome
WO2013150375A3 (en) * 2012-04-04 2014-01-09 Assistance Publique Hôpitaux De Paris Endothelial cells activation biomarkers characterizing antibody mediated rejection and uses thereof
US8932808B1 (en) 2004-01-21 2015-01-13 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for determining a graft tolerant phenotype in a subject
US8962261B2 (en) 2011-04-06 2015-02-24 The Board Of Trustees Of The Leland Stanford Junior University Autoantibody biomarkers for IGA nephropathy
US9290813B2 (en) 2009-12-02 2016-03-22 The Board Of Trustees Of The Leland Stanford Junior University Biomarkers for determining an allograft tolerant phenotype
US9535075B2 (en) 2010-03-25 2017-01-03 The Board Of Trustees Of The Leland Stanford Junior University Protein and gene biomarkers for rejection of organ transplants
US9803241B2 (en) 2008-08-18 2017-10-31 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for determining a graft tolerant phenotype in a subject
US9938579B2 (en) 2009-01-15 2018-04-10 The Board Of Trustees Of The Leland Stanford Junior University Biomarker panel for diagnosis and prediction of graft rejection
USRE46843E1 (en) 2005-03-14 2018-05-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for evaluating graft survival in a solid organ transplant recipient
USRE47057E1 (en) 2005-03-14 2018-09-25 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for evaluating graft survival in a solid organ transplant recipient

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2607455A1 (en) * 2005-04-22 2006-11-02 Morphotek, Inc. Antibodies with immune effector activity and that internalize in endosialin-positive cells
DK2137217T3 (en) 2007-04-05 2014-03-17 Morphotek Inc Methods of inhibiting the binding of endosialin to ligands
JP2012501456A (en) * 2008-08-28 2012-01-19 アスチュート メディカル,インコーポレイテッド Methods and compositions for diagnosis and prognosis of renal injury and renal failure
EP2813848A3 (en) * 2008-08-29 2015-03-11 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
AU2009308375B2 (en) * 2008-10-21 2015-06-25 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
EP3783363A1 (en) 2008-10-21 2021-02-24 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
CN104330574B (en) 2008-11-10 2017-04-12 阿斯图特医药公司 Methods and compositions for diagnosis and prognosis of renal injury and renal failure
JP5735922B2 (en) * 2008-11-22 2015-06-17 アスチュート メディカル,インコーポレイテッド Methods for the diagnosis and prognosis of kidney injury and renal failure
US9229010B2 (en) 2009-02-06 2016-01-05 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
BR112012002711A2 (en) 2009-08-07 2016-11-01 Astute Medical Inc method for assessing renal status in an individual, and protein medication
CA2779902A1 (en) 2009-11-07 2011-05-12 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
US20120156701A1 (en) * 2009-12-20 2012-06-21 Joseph Anderberg Methods and compositions for diagnosis and prognosis of renal injury and renal failure
JP5763098B2 (en) 2009-12-20 2015-08-12 アスチュート メディカル,インコーポレイテッド Methods and compositions for diagnosis and prognosis of renal injury and renal failure
NZ601590A (en) 2010-02-05 2014-10-31 Astute Medical Inc Methods and compositions for diagnosis and prognosis of renal injury and renal failure
JP5998057B2 (en) 2010-02-26 2016-11-30 アスチュート メディカル,インコーポレイテッド Methods and compositions for diagnosis and prognosis of kidney injury and renal failure
CA2804297A1 (en) 2010-06-23 2011-12-29 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
NZ605561A (en) 2010-06-23 2015-03-27 Astute Medical Inc Methods and compositions for diagnosis and prognosis of renal injury and renal failure
ES2933570T3 (en) 2011-12-08 2023-02-10 Astute Medical Inc Methods and compositions for the diagnosis and prognosis of kidney injury and kidney failure
EP3734280B8 (en) 2013-01-17 2022-08-24 Astute Medical, Inc. Methods and compositions for diagnosis and prognosis of renal injury and renal failure
WO2017214203A1 (en) 2016-06-06 2017-12-14 Astute Medical, Inc. Management of acute kidney injury using insulin-like growth factor-binding protein 7 and tissue inhibitor of metalloproteinase 2
KR102350228B1 (en) * 2019-06-21 2022-01-12 울산대학교 산학협력단 Urinary exosome-derived biomarkers for diagnosis or prognosis of T cell-mediated rejection in kidney allografts

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004018710A2 (en) * 2002-08-22 2004-03-04 Novartis Ag Diagnosis of chronic rejection
WO2004074815A2 (en) * 2003-02-14 2004-09-02 Beth Israel Deaconess Medical Center, Inc. Predicting graft rejection
WO2005054503A2 (en) * 2003-12-03 2005-06-16 Novartis Ag Biomarkers for graft rejection
US20050158721A1 (en) * 2002-01-16 2005-07-21 Shiels Paul G. Tissue rejection
WO2006125301A1 (en) * 2005-05-23 2006-11-30 The Governors Of The University Of Alberta Diagnostic applications of microarrays in organ transplantation

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5744101A (en) * 1989-06-07 1998-04-28 Affymax Technologies N.V. Photolabile nucleoside protecting groups
US5143854A (en) * 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US20020168358A1 (en) * 2001-04-30 2002-11-14 Gladue Ronald P. Treatment of T-cell mediated diseases
US7026121B1 (en) * 2001-06-08 2006-04-11 Expression Diagnostics, Inc. Methods and compositions for diagnosing and monitoring transplant rejection
US20060269948A1 (en) * 2005-05-16 2006-11-30 Halloran Philip F Tissue rejection

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050158721A1 (en) * 2002-01-16 2005-07-21 Shiels Paul G. Tissue rejection
WO2004018710A2 (en) * 2002-08-22 2004-03-04 Novartis Ag Diagnosis of chronic rejection
WO2004074815A2 (en) * 2003-02-14 2004-09-02 Beth Israel Deaconess Medical Center, Inc. Predicting graft rejection
WO2005054503A2 (en) * 2003-12-03 2005-06-16 Novartis Ag Biomarkers for graft rejection
WO2006125301A1 (en) * 2005-05-23 2006-11-30 The Governors Of The University Of Alberta Diagnostic applications of microarrays in organ transplantation

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
FLECHNER S.M. ET AL.: "Kidney transplant rejection and tissue injury by gene profiling of biopsies and peripheral blood lymphocytes", AMERICAN JOURNAL OF TRANSPLANTATION, vol. 4, no. 9, September 2004 (2004-09-01), pages 1475 - 1489, XP008103279 *
STEGALL M. ET AL.: "Gene expression during acute allograft rejection: novel statistical analysis of microarray data", AMERICAN JOURNAL OF TRANSPLANTATION, vol. 2, no. 10, November 2002 (2002-11-01), pages 913 - 925, XP003004281 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8932808B1 (en) 2004-01-21 2015-01-13 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for determining a graft tolerant phenotype in a subject
USRE46843E1 (en) 2005-03-14 2018-05-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for evaluating graft survival in a solid organ transplant recipient
USRE47057E1 (en) 2005-03-14 2018-09-25 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for evaluating graft survival in a solid organ transplant recipient
US9803241B2 (en) 2008-08-18 2017-10-31 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for determining a graft tolerant phenotype in a subject
US9938579B2 (en) 2009-01-15 2018-04-10 The Board Of Trustees Of The Leland Stanford Junior University Biomarker panel for diagnosis and prediction of graft rejection
US10538813B2 (en) 2009-01-15 2020-01-21 The Board Of Trustees Of The Leland Stanford Junior University Biomarker panel for diagnosis and prediction of graft rejection
WO2010096160A1 (en) * 2009-02-17 2010-08-26 The Board Of Trustees Of The Leland Stanford Junior University Compartment-specific non-hla targets for diagnosis and prediction of graft outcome
US9290813B2 (en) 2009-12-02 2016-03-22 The Board Of Trustees Of The Leland Stanford Junior University Biomarkers for determining an allograft tolerant phenotype
US10385397B2 (en) 2009-12-02 2019-08-20 The Board Of Trustees Of The Leland Stanford Junior University Biomarkers for determining an allograft tolerant phenotype
US9535075B2 (en) 2010-03-25 2017-01-03 The Board Of Trustees Of The Leland Stanford Junior University Protein and gene biomarkers for rejection of organ transplants
US11768208B2 (en) 2010-03-25 2023-09-26 The Board Of Trustees Of The Leland Stanford Junior University Protein and gene biomarkers for rejection of organ transplants
US8962261B2 (en) 2011-04-06 2015-02-24 The Board Of Trustees Of The Leland Stanford Junior University Autoantibody biomarkers for IGA nephropathy
WO2013150375A3 (en) * 2012-04-04 2014-01-09 Assistance Publique Hôpitaux De Paris Endothelial cells activation biomarkers characterizing antibody mediated rejection and uses thereof

Also Published As

Publication number Publication date
US20090176656A1 (en) 2009-07-09
EP2049713A1 (en) 2009-04-22
EP2049713A4 (en) 2010-06-16

Similar Documents

Publication Publication Date Title
EP2049713A1 (en) Tissue rejection
Reeve et al. Diagnosing rejection in renal transplants: a comparison of molecular-and histopathology-based approaches
US10538813B2 (en) Biomarker panel for diagnosis and prediction of graft rejection
Baron et al. Prediction of graft-versus-host disease in humans by donor gene-expression profiling
US7811767B2 (en) Methods and compositions for assessing acute rejection
Melk et al. Transcriptional analysis of the molecular basis of human kidney aging using cDNA microarray profiling
CA2553551C (en) Methods and compositions for determining a graft tolerant phenotype in a subject
US9746479B2 (en) Methods and compositions to predict and detect acute rejection
WO2006125301A1 (en) Diagnostic applications of microarrays in organ transplantation
US20110189680A1 (en) Methods of Diagnosing Rejection of a Kidney Allograft Using Genomic or Proteomic Expression Profiling
CA2605143A1 (en) Diagnosis of sepsis
CA2942384C (en) Method for identifying kidney allograft recipients at risk for chronic injury
WO2017100259A1 (en) Pretransplant prediction of post-transplant acute rejection
US20150191787A1 (en) Urine mrna profile and acute dysfunction of kidney allograft
US20100190166A1 (en) Tissue rejection biomarkers
US20100248251A1 (en) Tissue Rejection
Mandegary et al. Effect of Donor Tumor Necrosis Factor-[alpha] and Interleukin-10 Genotypes on Delayed Graft Function and Acute Rejection in Kidney Transplantation
US20100305038A1 (en) Diagnostic of immune graft tolerance using tmtc3 gene expression levels
Shangaris et al. Single-cell transcriptomics reveals markers of regulatory T cell dysfunction in Gestational Diabetes Mellitus
KR20240031236A (en) Non-invasive diagnosis of subclinical rejection
Einecke et al. CYTOTOXIC T CELLS, INTERFERON-GAMMA AND THE RENAL RESPONSE: PATHOGENESIS-BASED TRANSCRIPT SETS HAVE A HIGH DIAGNOSTIC VALUE IN HUMAN KIDNEY ALLOGRAFT REJECTION.
WO2022243388A1 (en) Molecular classifiers for antibody-mediated and t-cell mediated kidney transplant rejection
Yang et al. Endothelial-epithelial-related transcriptional levels in acute rejection biopsies of kidney transplant recipients are predictive for a worse response to steroid treatment
WO2009003273A1 (en) Assessing tissue rejection
McDaniel et al. Role of Biological Markers in Kidney Transplantation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07784962

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 12374639

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007784962

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: RU